• KATEDRA ZA ONKOLOGIJO • SEKCIJA ZA INTERNISTIČNO ONKOLOGIJO #2 INTERNATIONAL SUMMER SCHOOL IN MEDICAL ONCOLOGY ONKOLOŠKI INŠTITUT LJUBLJANA 7.-10. SEPTEMBER 2021 Strokovni odbor: prof. dr. Janja Ocvirk, dr.med. prof. dr. Davorin Radosavljević, dr.med. doc. dr. Tanja Mesti, dr.med. Organizacijski odbor: prof. dr. Janja Ocvirk, dr.med. doc. dr. Tanja Mesti, dr.med. Uredniki zbornika: Marko Boc, dr.med. prof. dr. Janja Ocvirk, dr.med. doc. dr. Tanja Mesti, dr.med. Recezenti: doc. dr. Tanja Mesti, dr.med. doc. dr. Erika Matos, dr.med. Organizator in izdajatelj (založnik): Onkološki inštitut Ljubljana Sekcija za internistično onkologijo Katedra za onkologijo Zborniki šol o melanoma in ostale publikacije s strokovnih dogodkov so dosegljivi na spletnih straneh Onkološkega inštituta: www.onko-i.si/publikacije-strokovnih-dogodkov-oi Ljubljana, september 2021 Tuesday, September 7 08.50-09.10 09.10-09.30 09.30-10.00 10.00-10.30 10.30-10.45 10.45-11.15 11.15-11.45 11.45-12.15 12.15-12.45 12.45-13.15 13.15-13.30 13.30-13.50 13.50-14.20 14.20-15.00 15.00-15.40 15.40-16.10 16.10-17.00 Registration of participants Satellite Symposium Abbott The importance of the palliativist and nutritionist (Moderators Mesti, Ocvirk) The palliative care – is early too early? (Ebert) The nutritional support – is fat bad? (Dobrila) Discussion Break Systemic treatment – the other side of the coin Side effects of immunotherapy and the management (Hribernik) Our experience – interesting cases (Žižek, Hribernik) Side effects of chemotherapy (including extravasation) and management (Ovčariček) Side effects of TKI and Management (Bokal) Car-T Neurological complications (Carpentier) Discussion Break Future perspectives – are we there yet Systemic treatment of the skin cancer – where we stand (Moderators Ocvirk, Mesti) Adjuvant and neoadjuvant systemic treatment of melanoma (Schadendorf) Systemic treatment for metastatic melanoma (Kandolf Sekulović) Our experience – interesting cases (Vid Čeplak Mencin, Mesti) Systemic treatment of non melanoma (Ocvirk) Our experience – interesting cases (Sever, Ocvirk) NGS - the new Superhero in Oncology (Mesti) Systemic treatment of the skin cancer – where we stand Interesting interactive cases and discussion Wednesday, September 8 08.40-9.00 09.00-09.30 09.30-10.00 10.00-10.30 10.30-11.00 11.00-11.30 11.30-12.00 12.00-12.15 Satellite Symposium Servier Future perspectives – are we there yet (Moderators Ocvirk, Mesti) Systemic treatment of the gastric cancer – where we stand (Boc) Our experience – interesting cases (Erman, Mesti) Systemic treatment of the biliary cancer – where we stand (Reberšek) Systemic treatment for the pancreatic cancer – are we going forward (Pašič) How to approach NET/NEC (Ignjatovič) Our experience – interesting cases (Leskovšek, Ocvirk) HCC – lots has been going on (Ocvirk) Our experience – interesting cases (Stefanovski, Ocvirk) Precision systemic treatment of colorectal cancer – can we understand the complexity (Pleština) Discussion 12.15-12.40 Break Future perspectives – are we there yet (Moderator Matos) 12.40-13.00 Enrichment of treatment at metastatic NSCLC with PD-L1 overexpression (PD-L1≥50) (Radisavljević) 13.00-13.20 Clinical choices at metastatic NSCLC without actionable oncogenic driver regardless of PD-L1 status (Jakopović) 13.20-14.00 Systemic treatment of head and neck cancer – what's new in the old (Grašič) Our experience – interesting cases (Plavc, Zupančič, Grašič) 14.00-14.40 Management of cancer of unknown primary in the molecular era (Matos) Our experience – interesting cases (Cankar, Matos) 14.40-15.10 Systemic treatment of Ewing sarcoma – where do we stand (Unk) Our experience – interesting cases (Sokolova, Unk) 15.10-15.30 Discussion and conclusion 15.30-15.50 Satellite Symposium Amgen Thursday, September 9 Future perspectives – are we there yet (Moderator Škrbinc) 08.40-09.00 Satellite Symposium BMS 09.00-09.30 Systemic treatment of prostate cancer – standards and prespectives (Belev) 09.30-10.00 Systemic treatment of RCC - standards and prespectives (Šeruga) 10.00-10.30 An approach to patient with cancer and kidney disease (Milanez) 10.30-11.00 The systemic treatment of the bladder cancer – is something new going on (Gnjidić) 11.00-11.30 Systemic treatment of germinal tumors – could it get better (Škrbinc) 11.30-12.00 Systemic treatment of gynecological tumors – standards and perspectives (Mandič) 12.00-12.15 Discussion 12.15-12.45 Break Future perspectives – are we there yet (Moderator Borštnar) 12.45-13.15 Early and locally advanced hormone dependant Breast cancer – where do we stand (Bešlija) 13.15-13.45 Metastatic breast cancer – standards and perspectives (Borštnar) 13.45-14.00 Discussion and conclusion 14.00-14.20 Satellite Symposium Pfizer 14.20.14.40 Satellite Symposium Eli Lilly Friday, September 10 Future perspectives – are we there yet (Moderators Jezeršek, Pahole, Rugelj) 09.00-09.20 Satellite Symposium Roche (09.20-13.00) Standards and perspectives in the systemic treatment of Lymphomas 09.20-09.40 Diffuse Large B cell Lymphoma (Jezeršek) 09.40-10.00 Marginal Zone Lymphoma (Miljković) 10.00-10.20 Follicular Lymphoma (Južnič Šetina) 10.20-11.00 Break 11.00-11:20 Mantle Cell Lymphoma (Jagodic) 11.20-11.40 11.40-12.00 12.00-12.30 12.30-13.00 13.00-13.30 T Cell Lymphomas (Pahole) Hodgkin lymphoma (Rugelj) Break Principles of Radiotherapy in Lymphomas (Zadravec Zaletel) Discussion and Conclusion KAZALO Ebert M.: The palliative care – is early too early?...................................................................................... 8 Dobrila-Dintijana R.: The nutritional support – is fat bad?......................................................................................... 17 Hribernik N.: Side effects of immunotherapy and the management………………………………………... 18 Žižek A., Hribernik N.: Our experience – interesting cases…………………………………………………………… 22 Ovčariček T.: Side effects of chemotherapy (including extravasation) and management…………………... 25 Bokal U.: Side effects of TKI and Management………………………………………………………... 42 Carpentier A.: Car-T Neurological complications…………………………………………………………… 49 Kandolf-Sekulović L.: Systemic treatment for metastatic melanoma………………………………………………... 60 Čeplak-Mencin V., Mesti T.: Our experience – interesting cases……………………………………………………………89 Simetić L., Blazičević K., Herceg D.: Patient with BRAF mutated metastatic melanoma …………………………………………...90 Ocvirk J.: Systemic treatment of non melanoma ………………………………………………………...92 Sever M., Ocvirk J.: Our experience – interesting cases ………………………………………………………….121 Mesti T.: NGS - the new Superhero in Oncology ……………………………………………………..134 Boc M.: Systemic treatment of the gastric cancer – where we stand …………………………………136 Erman A., Mesti T.: Our experience – interesting cases …………………………………………………………..139 Reberšek M.: Systemic treatment of the biliary cancer – where we stand …………………………………141 Ignjatović M.: How to approach NET/NEC ………………………………………………………………...142 Leskovšek K., Ocvirk J.: Our experience – interesting cases …………………………………………………………..148 Ocvirk J.: HCC – lots has been going on ……………………………………………………………….150 Stefanovski D., Ocvirk J.: Our experience – interesting cases …………………………………………………………..174 Radosavljević D.: Enrichment of treatment at metastatic NSCLC with PD-L1 overexpression (PD-L1≥50) … 180 Jakopović M.: Clinical choices at metastatic NSCLC without actionable oncogenic driver regardless of PDL1 status ……………………………………………………………………………………..191 Grašič-Kuhar C.: Systemic treatment of head and neck cancer – what's new in the old ………………………207 Zupančič T., Zakotnik B., Grašič-Kuhar C.: Our experience – interesting cases …………………………………………………………..209 Matos E.: Management of cancer of unknown primary in the molecular era ………………………….211 Unk M.: Systemic treatment of Ewing sarcoma – where do we stand ………………………………..223 Sokolova A, Unk M.: Our experience – interesting cases …………………………………………………………..231 Belev B.: Systemic treatment of prostate cancer – standards and prespectives ………………………..236 Šeruga B.: Systemic treatment of RCC - standards and prespectives …………………………………...243 Gnjidić M.: The systemic treatment of the bladder cancer – is something new going on ……………….256 Škrbinc B.: Systemic treatment of germinal tumors – could it get better (Škrbinc) ……………………. 271 Mandić A.: Systemic treatment of gynecological tumors – standards and perspectives (Mandič)……... 295 Bešlija S.: Early and locally advanced hormone dependant Breast cancer – where do we stand ………302 Borštnar S.: Metastatic breast cancer – standards and perspectives (Borštnar) …………………………. 324 Jezeršek-Novaković B.: Diffuse Large B cell Lymphoma ……………………………………………………………327 Miljković M.: Marginal Zone Lymphoma ………………………………………………………………….331 Južnič-Šetina T.: Follicular Lymphoma ………………………………………………………………………..335 Jagodic M.: Mantle Cell Lymphoma ……………………………………………………………………..346 Pahole J.: T Cell Lymphomas ………………………………………………………………………….347 Rugelj U.: Hodgkin lymphoma………………………………………………………………………….349 Zadravec-Zaletel L.: Principles of Radiotherapy in lymphomas …………………………………………………..350 2nd SUMMER SCHOOL IN MEDICAL ONCOLOGY PALLIATIVE CARE Is early too early? Maja Ebert Moltara, MD mebert@onko-i.si Head of a Department for Acute Palliative Care Department of Medical Oncology 7 - 10 September 2021, Ljubljana, Slovenia PALLIATIVE CARE Is early too early? EARLY PALLIATIVE CARE 2 8 6 BASIC QUESTIONS: WHAT? For WHO? WHO provides? WHERE? WHEN? WHY? 3 WHAT? WHO definition of palliative care Palliative care is an approach that improves the quality of life of patients and their families facing the problem associated with life-threatening illness, through the prevention and relief of suffering by means of early identification and impeccable assessment and treatment of pain and other problems, physical, psychosocial and spiritual. 9 COMPREHENSIVE PALLIATIVE CARE COMPREHENSIVE PALLIATIVE CARE • pain • dyspnea • nausea • vomiting • fatigue • fear • anger • anxiaty • depression PHYSICAL SYMPTOMs PSYCOLOGY SOCIAL SUPPORT SPIRITUAL SYMPTOMs SUPPORT • isolation • family dynamic • financial support • question about life/death • religion • hope 6 10 WHO provides and WHERE? All medical and non-medical members of teams in institutions where incurable patients are treated. Basic palliative care (80% patient): All levels of health system (hospitals, community health centre, at home, senior homes, hospicih...) All. Specialied palliative care (20%): Does not substitute basic palliative care, but it upgrade it for the patients with the most difficult and complex problems Specialized teams (acute palliative care departent, mobile PC team) EAPC: White Paper on standards and norms for hospice and palliative care in Europe 7 CONTINOUS PALLIATIVE CARE SENIOR HOMES REGIONAL HOSPITALS FAMILY DOCTOR AND DISTRIC NURSE PATIENT WITH A FAMILY EMERGANCY TEAM UNIVERSITY HOSPITALS PC MOBILE TEAM HOSPIC MOVEMENT 8 11 For WHO? CANCER CHRONIC HEART FAILURE COPB ELDERLY Murray, S. A et al. BMJ 2008;336:958-959 months, weeks SPECIFIC THERAPY + EARLY PALLIATIVE CARE LATE PALLIATIVE CARE days months CARE OF THE GRIEVING Months, years CARE OF THE DYING Performans status WHEN? DEATH 10 12 Conceptual framework newer term less stigma more hospital based wider range of services lower definitional clarity less volunteer involment SUPPORTIVE CARE PALLIATIVE CARE HOSPICE CARE SPECIFIC THERAPY no evidence of disease early stage disease (curable) advanced disease (incurable) older term more stigma more home based more focused services higher definitional clarity more volunteer involment bereavement Hui, D. et al. Concepts and definitions for “supportive care,” “best supportive care, ” “palliative care, ” and “hospice care” in the published literature, dictionaries, and textbooks. Support. Care Cancer 21, 659–685 (2013). WHEN? 12 13 EARLY PALLIATIVE CARE Temel, NEJM 2010 Ferell, J Pain Manag, 2015 Bakitas, JCO 2015 Higginson 2015 Murakami BMC Pall 2015 Bakitas, JCO 2013 14 14 PALLIATIVE CARE Is early too early? PALLIATIVE CARE CAN NOT BE TOO EARLY 15 1. DENIAL 2. PALLIPHOBIA PHASES OF DEVELOPMENT: 3. PALLILALIA 4. PALLIACTIVE 15 Hope is like the sun, which, as we journey toward it, casts the shadow of our burden behind us. THANK YOU!!! 2013 Mayo Foundation for Medical education an Research, Mayo Clin Proc. 2013; 88 (8):859865 16 The nutritional supportis fat bad? Prof. Renata Dobrila-Dintinjana, MD. PhD. Damir Vučinić, MD. Clinical Hospital Center Rijeka School of Medicine, University of Rijeka, Croatia • Monounsaturated fat, n-3 PUFA, and fat soluble vitamins may have a profound influence on the prevention and/or suppression of cancer, whereas saturated fat and n-6 PUFA may increase the risk of carcinogenesis • Monounsaturated fat and n-3 fatty acids should be preferred over animal fats and other vegetable fats in the diet • Malnutrition is an important issue in cancer patients, which should be appropriately managed by structured collaboration of MDT • Pharmacological approach (EPA/MA) to the treatment of anorexia cachexia syndrome in cancer patients significantly improves QoL and probably prolongs OS 17 IMMUNE-RELATED ADVERSE EVENTS OF IMMUNE CHECKPOINT INHIBITORS Nežka Hribernik, MD Institute of Oncology Ljubljana 2nd Summer School in Medical Oncology September 2021 Possible mechanisms underlying irAE Postow MA, et al. NEJM 2018 18 Martins F, et al. Nature Clinical Oncology Reviews, 2019 Fatal irAE (cont`d) • Risk of fatality: – Myocarditis 40% – Hypophysitis, adrenal insufficiency, colitis 2 - 5% – 10 – 17% for other irAEs – Older patients at higher risk (impared functional reserve, comorbidities) Wang el al. JAMA ONCO 2018 19 Management of irAEs Champiat S et al. Ann Oncol 2016 20 Special populations that are not presented in RCT Rzeniewicz el al. Ann Oncol 2021 TAKE-HOME MESSAGES! • MULTIDISCIPLINARY APPROACH – Baseline assessment – Ongoing assessment – PATIENT & PHYSICIAN EDUCATION – Management protocols – Collaboration with emergency departments, GPs, specialists, visiting nurses!! 21 Melanoma patient with multiple irAE - clinical case Side effects of immunotherapy and the management 2nd International Summer school Ana Žižek, dr.med Nežka Hribernik, dr.med. 1/ Female, 1960 ● Family history: mother had skin cancer ● Comorbidities: arterial hypertension ● Year 2012: presented with skin lesion on her left thigh, changing in colour – June 2012 – excision: melanoma, Clark IV, Breslow 2,4 mm, no ulceration – September 2012 – reexcision of tumor bed + SLNB (Institute of Oncology LJ) – pT3a N0 (0/1), R0, stage IIA – FU 2/ 22 January 2021 progression of disease (DFI: 9 years) January 2021: palpable resistance in her left thigh, loss of 6 kg in 1 month, nausea, back pain, night sweats ● ● Diagnostic procedure: – Lab: LDH 2.66 ukat/L, S-100 1.250 ug/L, CRP 160 mg/L – Bx of resistance: melanoma metastasis → BRAF V600E mutation – PET/CT: high metabolic uptake in the liver, subcutaneous tissue of left thigh, bones (Th 4,5,8,11, L1,5, sacrum, pubis, scapula, 10th and 5th rib) – CT of the head: solitary metastasis in basal ganglia (1 cm) without edema; no neurological symptoms 3/ Systemic treatment ● February 2021: started with comboIT ipilimumab 3 mg/kg + nivolumab 1 mg/kg - Supportive treatment: Zoledronic acid, vitamin D, CaCO3 - No radiotherapy (1 week after 1st infusion her back pain was gone) March 2021: increased appetite, no back pain, reduction in size of subcutaneous resistance in left thigh --> continues with 2nd infusion of nivo1/ipi3 ● –Side effects: pruritus grade II (Th: antihistamine) ●April 2021 – early time-point PET/CT (study QTA): PMR in most localisations, S100 normalized ● April 2021: 3rd infusion of nivo1/ipi3 4/ 23 Multiple irAE End od April 2021 - Regional hospital admission: hepatitis gr. III, diarrhoea gr. I, arthralgia gr. ● • ● ● ● Treatment: steroids i.v. (2mg/kg/d)+Pneumocystis prophylaxis, PPI In one week no more liquid stools and no more pain in her joints regular lab controls - liver transaminases decreased to gr. I Tapering of dexamethasone End of May 2021: worsening of sympthoms - skin rash around waist, fatigue, transaminitis gr. III, diarrhoea gr. I → again higher dose of steroids p.o. (2mg/kg/d) ● After improvement of lab and sympthoms, SLOW tapering of dexametasone over 5 weeks ● ACTH test! ● Permanently completed IT due to irAE ● 5/ ● June 2021: PET/CT: complete metabolic response of all metastatic localisations ● July 2021: normal AST/ALT, bili, S100, LDH, CRP ● August 2021: regular check-up, no signs of progression 6/ 24 Side effects of chemotherapy and the management Tanja Ovčariček Oncology institute Ljubljana, 2021 Knowing side effects of certain treatment is basic knowledge every oncologist should have!  a number of potential side effects  Side effects and long term sequelae of chemotherapy major source of concern for patients  Education of patients is important for adherence to cht treatment 25 Topoisomeras e inhibitors antimetabolites Alkylating agents taxanes Vinca alkaloids  Gastrointestinal epithelial cells  Bone marrow cells  Hair follicles Nurgali K, et al. Front.Pharmacol.2018 Timing of chemotherapy side effects Acute (minutes to hours/first days) Subacute During cht application: - allergic/infusion reaction - cardiac arrythmia - exstravasation Early (in hours after cht application): - nausea/vomiting - malaise - tumor lysis syndrome - phlebitis - cystitis After couple of days/week: - stomatisis, gastroenteritis - renal insufficiency - myelosuppression - peripheral neurotoxicity - Fatigue - alopecia 26 Late After weeks, months, years - nail changes - organ failure (heart failure) - infertility - teratogeneticy and cancerogeneticy GASTROINTESTINAL SIDE EFFECTS OF CHEMOTHERAPY CHEMOTHERAPY INDUCED NAUSEA AND VOMITING (CINV)  the most common and feared side-effects among the patients  compromise treatment outcomes What should oncologist know about CINV?  Timing of nausea  Emetogenic potential of chemotherapy/individual risk  Patophysiology of CINV and drugs used Roila F et al, Ann Oncol 2016 27 CHEMOTHERAPY INDUCED NAUSEA AND VOMITING (CINV)  in two phases, the acute and the delayed phase: Acute CINV: up to 24 h Delayed CINV: more than 24 h after chemotherapy administration Anticipatory CINV: before next cycle Cht administration Next cht cycle Roila F et al, Ann Oncol 2016 Emetic classification of chemotherapy agents High risk Moderate risk Low risk Minimal risk (>90 % patients) (30-90 % patients) (10-30% patients) (<10 % patients) Anthracycline/cyclophosphamide combinationb Carmustine Cisplatin Cyclophosphamide ≥1500 mg/m2 Dacarbazine Mechlorethamine Streptozocin Azacitidine Bendamustine Carboplatin Clofarabine Cyclophosphamide <1500 mg/m2 Cytarabine >1000 mg/m2 Daunorubicin Doxorubicin Epirubicin Idarubicin Ifosfamide Irinotecan Oxaliplatin Romidepsin Temozolomidec Thiotepad Trabectedin 28 Cabazitaxel Cytarabine ≤1000 mg/m2 Docetaxel Eribulin Etoposide 5-Fluorouracil Gemcitabine Ixabepilone Methotrexate Mitomycin Mitoxantrone Nab-paclitaxel Paclitaxel Pemetrexed Pegylated liposomal doxorubicin Topotecan Trastuzumabemtansine Vinflunine Bleomycin Busulfan 2Chlorodeoxyadenosi ne Cladribine Fludarabine Pixantrone Pralatrexate Vinblastine Vincristine Vinorelbine Roila et. Al. Ann Oncol 2016 Mechanism of antiemetic drugs: 1. 2. 3. 4. 5. 6. 5HT3 RA acute CINV NK1 RA delayed CINV D2 RA Multiple R CB1 Other Navari RM, et al. NEJM 2016 Antiemetic arsenal:       5HT3 RA: granisetron, palonosetron, ondasetron NK1 RA: aprepitant, fosaprepitant, netupitant D2 RA: metoklopramid Multiple R: deksamethasone, olanzepin CB1: canabinoids Other: benzodiazepines Navari RM, et al. NEJM 2016 29 Roila et. Al. Ann Oncol 2016 CLINICAL CASE  50 years old women with T3N0M0 urothelial bladder cancer, hematuria, no comorbidities  Treatment plan: neoadjuvant cisplatin-based chemotherapy  Worried about nausea and vomiting High risk (>90 % patients)  Acute CINV: NK1 RA (aprepitant 125 mg)+5HT3 RA (granisetron 1 mg)+deksamethason 12 mg  Delayed CINV: aprepitant 80 mg 2.3 D+deksamethason 8 mg 2.-4. d  Persisting N/V: breakthrough CINV: antiemetics with different mechanism of action: metoclopramide/olanzapine 30 MUCOSITIS  inflammatory and/or ulcerative lesions of the oral and/or gastrointestinal tract  anatomical distribution: oral mucositis (OM), gastrointestinal mucositis (GIM, diarrhea), and proctitis  the incidence of clinically significant mucositis has been reported to range from 40% (standard dose cht), 60–100% (high-dose cht)  Predisposing factors:  various mucotoxicity for different cht agents: 5-FU,methotrexate, irinotecan, cyclophosphamide, cisplatin, anthracyclines and taxanes  bolus infusion tends to be more toxic  in general, if a patient develops mucositis in the first cycle of treatment, the probability of the condition recurring in a subsequent cycle is high Stein et al, 2010, Kwon et al. 2016 ORAL MUCOSITIS .  erythema of the movable mucosa which progress to form painful ulcerations often covered by a pseudomembrane  may be associated with microbial colonization that may remain localized or become disseminated, especially in patients with severe neutropenia-SEPSIS!!  first signs appear shortly after administration and usually peak at about days 7–14, is usually self-limiting Stein et al, 2010, Kwon et al. 2016, Peterson DE et al, Ann Oncol 2015 31 MASCC and ESMO have developed guidelines whith strategies for managing oral mucositis  effective preventative and treatment strategies are lacking  Preventative meassurements: Oral health at the start of and during chemotherapy: general hygiene standards, dental care, normal saline and baking soda-non-alcochol mouthwashes, regular tooth brush, dietary and behavioral measures Peterson DE et al, Ann Oncol 2015 CHEMOTHERAPY INDUCED DIARRHEA (CID)  CID is potentially fatal (5%) due dehidration and electrolyte imbalances  CHT regimens CID: 5-fluorouracil and irinotecan are associated with rates of CID of up to 80% with one third of patients experiencing severe (grade 3 or 4) diarrhea, taxanes  CID:-uncomplicated (grade 1–2 with no complications) or complicated (grade 3–4 with one or more complicating signs or symptoms), -early onset (<24 h after administration, irinotecan) or late onset (>24 h after administration) -persistent (present for >4 weeks) or non-persistent (present for <4 weeks)  Treatment CID: 1. 2. 3. 4. Modification of diet and re-hydration Loperamide Ocreotide: loperamide refractory diarrhea (48 h), severe diarrhea Tincture of opium: may be considered as a second-line therapy for persistent and uncomplicated diarrhea Peterson DE et al, Ann Oncol 2015, Stein et al. Ther Adv Med Oncol 2010 32 CLINICAL CASE  54 years old patient on adjuvant chemotherapy for T3N1M0 BC  4. cycle of adjuvant chemotherapy, 1. application of docetaxel 100 mg/m2, prior cht applications without meaningful AE  Docetaxel was administreted with deksamethasone premedication according to standard scheme without acute toxic reaction  D7: confusion, sleepiness, unable to answer questions; afebrile, RR 100/70, p: 100/min, normal ECG, without major neurologic deficit on examination, physical examination otherwise unrenarkabe  Heteroanamnesis: profound diarrhea (6-10 stools per day 5 days, last 2 days vomiting) without fever  Lab. Test: profound hyponatremia (Na: 115, K:2.1 mmol/l) elevated creatinin value and urea, CRP, PCT normal, no signs of myelosuppression  X-ray abdomen and stool cultures: negative  DIAGNOSIS: acute dehydration and hypovolemic hyponatriemia as a consequence of diarrhea, nausea and vomiting  Treated with parenteral saline infusion, loperamide  Adjuvant chemotherapy: weekly paclitaxel administration CHEMOTHERAPY INDUCED MYELOSUPPRESSION MYELOSUPPRESSION Leucopenia/ Neutropenia Thrombocytopenia Anemia 33 FEBRILE NEUTROPENIA DEFINITION:an oral temperature of >38.3°C or two consecutive readings of >38.0°C for 2 h and an absolute neutrophil count (ANC) of <0.5 × 109/l  FN remains one of the most frequent and serious complications of cancer chemotherapy  20-30 % pts require hospitalisation, mortality 10%  Predisposing factors: certain chemotherapeutics regimen-FN risk, age, advanced disease, prior FN, mucositis, low PS, CVS disease TREATMENT: 1. Urgent recognition 2. Immediate treatment PREVENTION: Chemoprpphylaxis according to risk for FN (individual, cht scheme) Klastersky J et al, Ann Oncol 2016 Klastersky J et al, Ann Oncol 2016 34 FEVER AND SUSPECTED NEUTROPENIA History, previous microbiology reports, physical examinationsource of infection? Lab test: blood counts, CRP, liver, kidney Blood cultures: i.v.cathethers, peripheral vein Urinanalysis X-ray Urin/sputum/other culture in case of suspicion ≥21 Low risk MONITORING!!! No pneumonia, stable, no i.v. cathethers: Po/iv antibiotics: amoksicillin plus clavulanic acid and quinolones Early discharge? • extravasation • alopecia • nail changes • hand-foot syndrome (capecitabin, taxanes, 5-FU) • dermatitis 35 <21 High risk i.v. broad spectrum antibiotics Klastersky J, eta al. Ann Oncol 2016  HFS:palmar-plantar erythrodysaesthesia syndrome: redness, marked discomfort, swelling and tingling in the palms of the hands or the soles of the feet (5-fluorouracil (5-FU), (6%-34%), capecitabine (50%-60%), doxorubicin (22%-29%), PEGylated liposomal doxorubicin (40%-50%), docetaxel [6%-58%; preventative mesures are important-10% urea cream significantly reduces the incidence of HFS  Treatment of HFS: , skin inflammation: high-potency topical corticosteroids], while erosions and ulcerations: with antiseptic solutions (silver sulfadiazine 1%), analgesia on painful areas: lidocaine 5% patches  Skin cooling (e.g. cold gloves or socks) significantly reduce of HFS for ChT given as an infusion( paclitaxel, docetaxel and liposomal doxorubicin) Lacouture ME, et al, Ann Oncol 2020 EXTRAVASATION:inadvertent infiltration of chemotherapy into the tissues surrounding the intravenous site  Extravasated drugs are classified according to their potential for causing damage as ‘vesicant’, ‘irritant’ and ‘nonvesicant’ VESICANT: blisters/tissue destruction !!!!! IRRITANTS: pain along the vein and inflammatory reaction . DNA-binding vesicant drugs Mechloretamine Doxorubicin, Daunorubicin, Epirubicin, Idarubicin Mitomycin, Dactinomycin, Mitoxantrone* Alkylating agents Anthracyclines Antitumor antibiotics Non-DNA binding vesicant drugs Vinca alkaloids Taxane Vinblastine, Vincristine, Vinorelbine, Vindesine Paclitaxel, Docetaxel Irritant drugs Alkylating agents Platinum analogs Topoisomerase II inhibitors Anthracyclines Topoisomerase I inhibitors Carmustine, Dacarbazine, Ifosfamide, Melphalan, Thiotepa, Carboplatin,° Cisplatin,# Oxaliplatin Teniposide, Etoposide Liposomal Doxorubicin, Liposomal Daunorubicin Irinotecan, Topotecan 36 Fidalgo JA, et al, Ann Oncol 2012 Extravasated vesicant Anthracyclines Antidote Treatment DMSO Dexrazoxane Topical cooling Docetaxel Hyaluronidase Topical cooling Mytomicin DMSO Topical cooling Mitoxantrone DMSO Topical cooling Vinca alkaloids Hyaluronidase Topical warming  DMSO (topical application): is a solvent capable of penetrating tissue, it neutralizes free-radical accumulation and encances systemic absorption of the extravasated drug.  Dexrazosane (intravenously): is approved for anthracycline extravasation treatment, it binds to iron and prevents the formation of free radicals which induce extravasation-induced tissue necrosis.  Hyaluronidase (subcutaneously) degrades hyaluronic acid, breaks down subcutaneous tissue bonds promoting drug diffusion and enhances the absorption of injected substances. Signs and symptoms of vesicant extravasation: swelling, redness and/or discomfort, resistance during drug administration, a slow and sluggish infusion, and lack or loss of a blood return from the i.v. cannula, implanted port or other central venous access device. Fidalgo JA, et al, Ann Oncol 2012 It is essential to recognize that an extravasation has taken place as quickly as possible!!!  40 years BC patients develops sweeling and redness in the arm where the canula during doksorubicin infusion is inserted WHAT WOULD YOU DO? Fidalgo JA, et al, Ann Oncol 2012 37 CENTRAL AND PERIPHERAL CHEMOTHERAPY INDUCED NEUROTOXICITY (CIPN)  chemotherapy-induced cognitive dysfunctions=„chemo-brain“, other: other types of acute encephalopathy: ifosfamide-induced acute encephalopathy, increased risk of trombemolic stroke  incidence of CIPN approximately 38% (up to 90% with oxaliplatin)  reduce functional capacity and quality of life, the long-term reversibility questionable, symptoms may last years after chemotherapy discontinuation  CIPN: platinum-based agents (cisplatin, oxaliplatin), vinca alkaloids (vincristine, vinorelbine), taxanes (docetaxel, paklitaksel)  Oxaliplatin-Induced Peripheral Neuropathy:  more than 90% of patients developed acute neuropathy (paresthesia, dysesthesia of the hands, feet and perioral area induced by cold stimuli in the hours and days ) and 30–50% of patients developed chronic neuropathy (paresthesia, numbness, sensory ataxia )  Taxane, vinca alkaloids neuropathy: sensory neuropathy with a stocking-and-glove distribution over the hands and feet Kerckhove N, et al. F Phar 2017 Kerckhove N, et al. F Phar 2017 38  no effective agent exists to prevent CIPN  prevention of CIPN with cryotherapy can be considered according to ESMO, similar recommendation for medical exercise for cancer patients (low LoeE/GoR-IIC)  Management of CIPN:- reducing or discontinuing chemotherapy when CIPN develops - treatment of the symptoms of neuropathic pain- only duloxetine was shown to help neuropathic pain in established CIPN; any other medications (gabapentin, topical preparations, etc.) are used in an off-label fashion Jordan B, et al. Ann Oncol 2020 True allergic responses vs non-allergic responses Rosello S, et al, Ann Oncol 2016 39 Rosello S, et al, Ann Oncol 2016 CARDIOTOXICITY Arrythmias: taxanes Coronary artery spasem:5-FU, capecitabin, cisplatin Heart failure: anthracyclines 40  Use all the knowledge for preventative meassures!  Educate the patient! THANK YOU FOR YOUR ATTENTION! 41 Toxicity of tyrosine kinase inhibitors (TKI) and the management Urška Bokal 2nd Summer School of Medical Oncology, 7/ 9/2021 Tyrosine kinase inhibitors Tyrosine kinases: • activate proteins/autoactivate by phosphorylation of tyrosine moiety important for signal transduction and cell cycle regulation Tyrosine kinase inhibitors: • Small molecules, oral application • act mostly by blocking ATP binding site, therefore inhibit phosphorylation • bind reversibly or irreversibly ATC classification system Other protein kinases: B Raf (serine threonine kinase) https://www.whocc.no/atc_ddd_index/?code=L01XE&showdescription=no 42 On- and off- target toxicity On-target: • due to inhibition of the desired target (mechanism based) • class effect: shared with all agents that inhibit specific target • VEGFR TKI: hypertension • EGFR TKI: rash Off-target: • due to inhibiton of other unintended targets • „off targets“ share structures or residues with the intended targets • sunitib: hematologic toxicity (FLT3 inhibition) • toxicities can ovelap due to cross interaction of multiple pathways CA Cancer J Clin. 2013;63:249-79 https://www.targetedonc.com/publications/targeted-therapies-cancer/2013/december-2013/Toxicities-of-Targeted-Therapies-and-Their-Management The good news: toxicity may correlate with response/better survival • rash due to EGFR TKI in lung cancer • hypertension and hypothyroidism due to VEGFR TKI in renal cell carcinoma Liu S et al. Cancer Treat Rev. 2014; 40: 883-91 43 ErbB tyrosine kinase inhibitors Shah R et al. Drug Safety. 2019; 42:181-98 all agents: hepatotoxicity spc of selected drugs ErbB TKI toxicity: management • drug interruption, lowering of the dose • life threatening: discontinuation of treatment Up to Date Shah R et al. Drug Safety. 2019; 42:181-98 Lacouture M et al. Am J Clin Dermatol. 2018; 19: S31-9 44 VEGFR tyrosine kinase inhibitors VEGFR TKI – management of cardiovascular toxicity CA Cancer J Clin. 2013;63:249-79 TE: thromboembolic events Up to Date 45 VEGFR TKI – management of toxicity CA Cancer J Clin. 2013;63:249-79 Rimassa L et al. Can Treat Rev 2019; 77: 20-8 ALK tyrosine kinase inhibitors CPK – creatine phosphokinase AP – alkaine phosphatase ALL: interstital lung disease Kassem L et al. Crit Rev Oncol Hematol 2019; 134:56-64 smpc 46 ALK TKI – management of toxicity Kassem L et al. Crit Rev Oncol Hematol 2019; 134:56-64 smpc http://www.bccancer.bc.ca/drug-database-site/Drug%20Index/Crizotinib_monograph_1Sep2014.pdf MEK 1,2 inhibitors (+ B-RAF inhibitors) CK – creatine phosphokinase MEK1,2 inhibitors: • ↓ LVEF, hypertension, • retinal pigment epithelial detachment/retinal vein occlusion, • interstitial lung disease, B-RAF inhibitors: • QT prolongation (vemurafenib, encorafenib), • uveitis, • cutaneus squamous cell carcinoma, keratoacantomas, hyperkeratosis, • pyrexia Kdaud A et al. The Oncologist 2017;22:823-33 smpc 47 Ocular toxicity Possible mechanism of MET TKI ocular toxicity: • ERK activation is important for photoreceptor survival • MEK inhibition results in apoptosis and loss of differentiation during photoreceptor development EGFR TKI: keratoconjunctivitis crizotinib: poor light-dark adaptation Kdaud A et al. The Oncologist 2017;22:823-33 smpc Take home messages • Toxicity varies between patients. • Beware of interactions with food and drugs! • Multidisciplinary management: referral to doctors of other specialities. • Chronic low grade toxicity influence the quality of life of patients. 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 SURVIVAL ANALYSIS IN SLOVENIAN PATIENTS WITH METASTATIC MELANOMA AND IMMUNE RELATED ADVERSE EVENTS Vid Čeplak Mencin assist. prof. Tanja Mesti, MD, PhD Summer School 2021 Institute of Oncology Ljubljana, sept. 2021 CONCLUSION • irAE cohort: better treatment outcome, longer time to disease progression, better ORR • ORR: irAE 57%, NirAE 37% • PFS: irAE 301,6 days, NirAE 247,3 days • SP: irAE 80%, NirAE 60% • Better SP in patients with elevated LDH and irAE M1a/b (TNM classification) • Worse SP for M1c/d – new type of melanoma? 89 Patient with BRAF mutated metastatic melanoma Sequencing, rechallenge, new therapies... L.Simetic, K.Blazicevic and D. Herceg On behalf of MDT for melanoma and skin cancers UHC Zagreb, Croatia QUESTIONS • Should we continue with BRAF MEK therapy or not? Patient was treated with 2 lines in metastatic setting so no other option, except Cht, is available in Croatia ( by National regulatory agency for health insurance) • Do we have „ a joker card” with previous response to ICI ( lung mets regression) ? Rechallenge with mono PD-1 inhibitors or combo ICI? • Clinical study/ compassionate use programe:: ICI+ lenvatinib or? • Something new like Fecal Transplant therapy + ICI? • Your comments are precious  90 Multidisciplinary team for melanoma treatment University hospital centre Zagreb; core members  dermatologist, dermatooncologist: D. Štulhofer (chair), R.Čeović  plastic surgeon: D. Mijatović, S. Smuđ Orehovec  head and neck surgeon: M.Jurlina, J.Biloš, D. Leović  ophthalmologist: N.Vukojević, M. Štanfel  medical oncologist: D.Herceg, K.Blažičević, L.Simetić  radiooncologist: F.Šantek  pathologist: S.Dotlić, I.Ilić  nuclear medicine specialist: G.Horvatić Herceg, S. Kusačić Kuna, M.Ciglar  radiologist: M.Lušić, M.Kralik lsimetic@kbc-zagreb.hr 91 Systemic treatment of nemelanomskih skin cancer Prof. Janja Ocvirk Ljubljana, 7.9.2021 Systemic treatment of non-melanoma skin cancers - immunotherapy? 92 Squamous cell carcinoma of the skin The second most common NMSC (20%) The incidence has been growing over the last 30 years(50-200%) Head and neck 80-90% 90% has a good prognosis What about the rest 10%? SCC in transplant patients 36 times higher incidence than usual(BCC: SCC 4: 1) Aggressive course - poor prognosis 93 Tumor Mutational Burden in CSCC Formation of Neoantigens Somatic Mutation Frequency (/Mb) 1,000 100 61.2 8.2 10 Potential Sensitivity to Immunotherapy Frequently 13.2 3.2 Regularly 1 0.1 Occasionally 0.01 SCCHN (178) LUSC (178) Melanoma (121) CSCC (39) Tumor Type Red horizontal line and associated number in figurer = median mutations per MB. CSCC, cutaneous squamous cell carcinoma; LUSC, lung squamous cell carcinoma; Mb, megabase of DNA; SCCHN, Squamous cell carcinoma of the head and neck. Pickering CR, et al. Clin Cancer Res. 2014;20:6582–6592. Tumour mutational burden & objective response rate Anti–PD-1 and anti–PD-L1 therapy across 27 tumour types1 1. Yarchoan M, et al. N Engl J Med. 2017; 377: 2500–01; 2. Papadopoulos K, et al. Presentation at 2017 American Society of Clinical Oncology, Chicago, IL. J Clin Oncol. 2017;35(15_suppl):9503; Adapted from Yarchoan et al, 2017 94 Reasons for immunotherapy in CSCC High tumor mutation load (TMB) and immunogenic cancer High TMB may contribute to increased neoantigen production, which may increase tumor antigenicity 1 Immunosuppression is a well-described risk factor for CSCC (especially in organ transplant patients)2 PD-L1 expression was detected in advanced CSCC3 1. Pickering CR, et al. Clin Cancer Res. 2014;20:6582-92; 2. Euvrard E, et al. N Engl J Med. 2003;348:1681-1691. 3. Slater NA, et al. J Cutan Pathol. 2016;43:663-70. Candidates for immunotherapy in advanced CSCC Patients with advanced CSCC Locally advanced / metastatic disease Patients with recurrences after previous surgeries Patients who are not candidates for surgery due to morbidity / potential exhaustion or a low level of confidence within clear boundaries Patients who are not candidates for radiotherapy 95 Migden MR, et al. N Engl J Med. 2018;379:341-351. 96 EMPOWER-CSCC-1 study design 1 Metastatic CSCC (nodal and/or distant) 2 Locally advanced CSCC Key inclusion criteria Cemiplimab 3 mg/kg Q2W IV* for up to 96 weeks • • • Tumour imaging every 8 weeks for the assessment of efficacy (retreatment optional for patients with progression during follow-up†) • ECOG performance status of 0 or 1 Adequate organ function At least one lesion measurable by RECIST 1.1 criteria (for scans) or modified WHO criteria (for photos) CSCC lesion that is not amenable to surgery or radiation therapy per investigator assessment Key exclusion criteria Metastatic CSCC (nodal and/or distant) 3 Tumour imaging every 9 weeks for the assessment of efficacy Cemiplimab 350 mg Q3W IV for up to 54 weeks • • • Tumour response assessment by ICR (RECIST 1.1 for scans: modified WHO criteria for photographs) Ongoing or recent (within 5 years) autoimmune disease requiring systemic immunosuppression Prior anti–PD-1 or anti–PD-L1 therapy History of solid organ transplant, concurrent malignancies (unless indolent or not considered life threatening; for example, basal cell carcinoma), or haematologic malignancies †To account for possible pseudoprogression, treatment could be continued beyond initial RECIST 1.1-defined progression informed by ir-related response criteria *Cemiplimab is not licensed at this dose for the treatment of CSCC CSCC, cutaneous squamous-cell carcinoma; ECOG, Eastern Cooperative Oncology Group; ICR, independent central review; IV, intravenous; PD-1, programmed cell death-1; PD-L1, programmed death-ligand 1; Q2W,every 2 weeks; Q3W, every 3 weeks; RECIST, Response Evaluation Criteria in Solid Tumours; WHO, World Health Organisation. Migden MR, et al. Presentation at 2019 American Society of Clinical Oncology, Chicago, IL. J Clin Oncol. 2019;37(15_suppl):6015 CSCC, cutaneous squamous cell carcinoma 1 1 EMPOWER-CSCC-1 treatment arms Phase II data supporting cemiplimab license (n=193) Group 1 Group 2 Group 3 Metastatic Locally advanced Metastatic Number of patients 59 78 56 Cemiplimab dosing 3 mg/kg Q2W 3 mg/kg Q2W 350 mg Q2W Sept 20 2018 Oct 10 2018 Sept 20 2018 16.5 9.3 8.1 CSCC Data cut-off Median follow-up (months) Cemiplimab SmPC, available at https://www.ema.europa.eu/en/documents/product-information/libtayo-epar-product-information_hr.pdf; accessed January 2020 97 EMPOWER-CSCC-1 Response rates in three treatment groups Group 1 Group 2 Group 3 Metastatic CSCC Cemiplimab 3 mg/kg Q2W* N=59 Locally advanced CSCC Cemiplimab 3 mg/kg Q2W* N=78 Metastatic CSCC Cemiplimab 350 mg Q3W N=56 49.2% 35.9, 62.5 43.6% 32.4, 55.3 39.3% 26.5, 53.2 16.9% 12.8% 3.6% Objective response rate 95% Confidence interval Confirmed objective response Complete response† Partial response 32.2% 30.8% 35.7% Stable disease 15.3% 35.9% 14.3% Progressive disease 16.9% 11.5% is not licensed at this dose for 26.8% *Cemiplimab the treatment of CSCC †Only patients with complete healing of prior cutaneous involvement; for locally advanced CSCC, biopsy required to confirm complete response CSCC, cutaneous squamous-cell carcinoma; Q2W, every 2 weeks Cemiplimab SmPC, available at https://www.ema.europa.eu/en/documents/product-information/libtayo-epar-product-information_hr.pdf; accessed January 2020 Kaplan–Meier Estimation Overall Survival, Progression-Free Survival, and Duration of Response in Advanced CSCC Patients 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 Median PFS by ICR was 18.4 months (95% CI: 7.3– not evaluable) 0 2 4 6 Probability of survival Probability of PFS Metastatic CSCC (Group 1)1 8 10 12 14 16 18 20 22 24 26 28 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 Median OS has not been reached; Kaplan-Meier estimation of OS at 24 months was 70.6% (95% CI: 57.0–80.6;) 0 2 4 6 Number 59 43 39 36 32 26 26 26 25 18 15 10 at risk 8 10 12 14 16 18 20 22 24 26 28 Months Months 1 0 0 Number 59 56 52 49 47 47 46 41 39 32 24 14 6 at risk Median DOR not reached Locally Advanced CSCC (Group 2)2 Median PFS K-M Estimated PFS at 12 months Median OS K-M Estimated OS at 12 months Median DOR NR 58.1% (95% CI: 43.7–70.0) NR 93.2% (95% CI: 84.4–97.1) NR Group 1: Median duration of follow-up = 16.5 mos (range 1.1 – 26.6); Group 2: Median duration of follow-up = 9.3 mos (range 0.8 – 27.9) Data cut-off date: Sept 20, 2018 (Group 1); Oct 10, 2018 (Group 2) CI, confidence interval; CSCC, cutaneous squamous cell carcinoma; ICR, independent central review; OS, overall survival; PFS, progression-free survival; NR, not reached 1. Guminski et al. J Clin Oncol. 2019:37 (suppl; abstr 9526) [poster presentation]. 2. Migden MR, et al. J Clin Oncol. 2019:37 (suppl; abstr 6015) [poster presentation]. 98 1 0 Probability of no progression or death EMPOWER-CSCC-1:Duration of response K-M estimated event-free probability by ICR in responding patients Data cut-off date: 20 Sep 2018 (Group 1); 10 Oct 2018 (Group 2). 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 88.9% 87.8% 96.4% 96.2% 82.5% 87.8% Metastatic CSCC Locally advanced CSCC 0 Number of patients at risk Metastatic CSCC 29 Locally advanced CSCC 2 4 6 8 10 12 14 Month 16 18 20 22 24 26 29 34 28 30 27 27 22 23 22 17 22 14 15 9 11 6 7 6 0 4 0 1 0 1 22 12 0 Median duration of response has not been reached. CSCC, cutaneous squamous cell carcinoma; ICR, independent central review. Cemiplimab adverse reaction profile Safety population N=591 Permanent discontinuation Serious adverse events 8.6% 5.8% Immune-related adverse reactions 20.1% 4.4% ≥Grade 3 irARs Grade 3 6.1% Grade 4 1.2% Grade 5 0.7% Most common irARs Hypothyroidism 7.1% 0 Pneumonitis 3.7% 1.9% Immune-related skin ARs 2.0% 0.3% Hyperthyroidism 1.9% 0 Hepatitis 1.9% 0.8% 9.1% 0.3% Infusion-related reactions Cemiplimab SmPC, available at https://www.ema.europa.eu/en/documents/product-information/libtayo-epar-product-information_hr.pdf; accessed January 2020 irARs, immune-related adverse reactions 99 Pembrolizumab for Recurrent/Metastatic Cutaneous Squamous Cell Carcinoma: Efficacy and Safety Results From Phase 2 KEYNOTE-629 Study Studiendesign CR, complete response; cSCC, cutaneous squamous cell carcinoma; DCR, disease control rate; DOR, duration of response; ECOG PS, Eastern Cooperative Oncology Group performance status LA, locally advanced; ORR, objective response rate; OS, overall survival; PFS, progression-free survival Q3W, every 3 weeks; R/M recurrent and/or metastatic. aPatients who discontinue treatment after achieving CR may be eligible to receive an additional 17 cycles of pembrolizumab if disease progression occurs. J.-J. Grob et al., KEYNOTE-629 Efficacy and Safety of pembrolizumab in patients with R/M cSCC, Poster presented at ESMO 2019 Pembrolizumab for Recurrent/Metastatic Cutaneous Squamous Cell Carcinoma: Efficacy and Safety Results From Phase 2 KEYNOTE-629 Study PFSa in the R/M Cohort OSa in the R/M Cohort NR, not reached; OS, overall survival; PFS, progression-free survival; R/M, recurrent and/or metastatic. aFrom product-limit (Kaplan-Meier) method for censored data. J.-J. Grob et al., KEYNOTE-629 Efficacy and Safety of pembrolizumab in patients with R/M cSCC, Poster presented at ESMO 2019 100 Cemiplimab is indicated as monotherapy in adult patients with metastatic or locally advanced squamous cell carcinoma of the skin who are not candidates for curative surgery or curative first-line radiation. 101 Basal cell carcinoma of the skin A major cause of BCC is exposure to UV radiation A major cause of BCC is exposure to UV radiation, leading to cumulative DNA damage and gene mutations1–5 Most sporadic cases of BCC arise from chronic sun-exposure1,2 Epidemiological data suggest the overall incidence of BCC is increasing significantly and show marked geographical variation1,6–8 Australia has the highest incidence rate of BCC in the world, reporting a rate of 1–2% per year1,6 80% occur on the head and neck 15% occur on the trunk 5% occur on the arms, legs or other sites 1. Rubin AI et al. N Engl J Med 2005;353:2262–9 2. Wong CSM et al. Br Med J 2003;327:794–8 3. Roewert-Huber J et al. Br J Dermatol 2007;157:47–51 4. Lear JT et al. J R Soc Med 1998;91:585–8 102 5. Caro I, Low JA. Clin Cancer Res 2010;16:3335–9 6. Diepgen TL, Mahler V. Br J Dermatol 2002;146(suppl):1–6 7. Ting PT et al. J Cutan Med Surg 2005;9:10–15 8. Rogers HW et al. Arch Dermatol 2010;146:283–7 Advanced basal cell carcinoma Locally advanced basal cell carcinoma (lnBCC) Aggressive disease with local tissue damage Frequent recurrences after surgery The operation would cause deformation BCC Metastatc BCC (mBCC) Locally advanced BCC Metastatski BCK nBCC (1-2%) Rare but serious form of BCK It involves the presence of metastases (e.g., lymph nodes, bones, lungs, liver 1 Weak outcome (median survival: 8-14 months2-3 5-year survival rate: 10% 3,4 1. Ting PT et al. J Cutan Med Surg 2005;9:10–15 2. von Domarus H, Stevens PJ. J Am Acad Dermatol 1984;10:1043–60 3. Lo JS et al. J Am Acad Dermatol 1991;24:715–19 4. Wong CSM et al. Br Med J 2003;327:794–8 23 Treatment of basal cell carcinoma Curettage and cavertisation, cryosurgery Cream imiquimod Surgical excision Electrochemotherapy nBCC Radiotherapy Targeted therapy – HHI:Vismodegib, Sonidegib, Immunotherapy 24 103 BCC and Hedgehog signal pathway 26 The pathway of cell growth and differentiation that controls the formation of organs in embryonic development • • The Hedgehog signaling pathway is inactive in most of the tissue of the adult Abnormal activation (mutation) of the Hedgehog signal pathway plays an important role in pathogenesis BCC1 Hedgehog signaling pathway inhibitors provide a new treatment option for advanced patients BCC (vismodegib, sonidegib) 104 105 RR in 1st line treatment - HHI 106 Phase 2 Study of Cemiplimab in Advanced BCC 31 (Study 1620) ‒ NCT031326361-3 An open-label, non-randomized Phase 2 study of cemiplimab in patients with advanced BCC who experienced progression on or intolerance to hedgehog pathway inhibitor therapy1-4 Group 1 Adult patients with metastatic (nodal and distant) BCC N=54 350 mg IV Q3W for up to 93 weeks (or until disease progression, unacceptable toxicity, or withdrawal of consent) Group 2 Adult patients with laBCC N=84 Primary endpoint • ORR by ICR Select secondary endpoint Cemiplimab Tumor response assessments: Q9W for treatment cycles 1-5, Q12W for treatment cycles 6-9* • DOR, PFS, OS, complete response by ICR, ORR per investigator, and safety and tolerability *Tumor response assessment by ICR (RECIST 1.1 and/or modified WHO criteria). BCC=basal cell carcinoma; DOR=duration of response; ICR=independent central review; IV=intravenous; laBCC=locally advanced BCC; N=number of patients; ORR=overall response rate; OS=overall survival; PFS=progression-free survival; Q3W=every 3 weeks; Q9W=every 9 weeks; Q12W=every 12 weeks; RECIST=Response Evaluation Criteria in Solid Tumors; WHO=World Health Organization. 1. ClinicalTrials.gov/NCT03132636. https://clinicaltrials.gov/ct2/show/NCT03132636. Accessed October 2020. 2. Stratigos AJ, et al. Poster presented at: European Society for Medical Oncology (ESMO) Virtual Congress; September 19–21, 2020:LBA47. 3. Lewis KD, et al. Poster presented at: Society for Immunotherapy of Cancer (SITC) Virtual Congress; November 9–14, 2020:Poster 428. Patient Eligibility Select Exclusion Criteria1-3 Select Inclusion Criteria1-3 Ongoing or recent (within 5 years) autoimmune disease requiring systemic immunosuppression • Adults (≥18 years) with histologically confirmed diagnosis of invasive BCC • • Prior disease progression on HHI therapy, or intolerance to prior HHI therapy,* or no better than stable disease after 9 months on HHI therapy • Prior anti–PD-1 or anti–PD-L1 therapy • Active brain metastases • Immunosuppressive doses of steroids (>10 mg prednisone daily or equivalent) • Concurrent malignancy other than BCC and/or history of malignancy other than BCC within 3 years of date of first planned dose of cemiplimab, except for tumors with negligible risk of metastasis • ≥1 measurable baseline lesion • ECOG performance status ≤1 • Adequate organ function • Must not be a candidate for radiation therapy or surgery *Defined as any Grade 3/4 HHI-related AEs, or any of the following Grade 2 HHI-related AEs following ≥3 months of exposure: muscle spasms or myalgias, dysgeusia or anorexia (if accompanied by Grade ≥1 weight loss), nausea, or diarrhea (despite medical management). BCC=basal cell carcinoma; ECOG=Eastern Cooperative Oncology Group; HHI=hedgehog inhibitor; PD-1=programmed cell death protein-1; PD-L1=programmed death-ligand 1. 1. ClinicalTrials.gov/NCT03132636. https://clinicaltrials.gov/ct2/show/NCT03132636. Accessed October 2020. 2. Stratigos AJ, et al. Poster presented at: European Society for Medical Oncology (ESMO) Virtual Congress; September 19–21, 2020:LBA47; 3. Lewis KD, et al. Poster presented at: Society for Immunotherapy of Cancer (SITC) Virtual Congress; November 9–14, 2020:Poster 428. 107 Kaplan-Meier Curve for PFS by ICR in Patients with 33 Progression-free Survival laBCC 1.0 0.9 Probability of PFS 0.8 0.7 0.6 0.5 0.4 0.3 0.2 Estimated median PFS, months: 19.3[95% CI, 8.6–NE] 0.1 Estimated 12-month event-free probability: 56.5% [95% CI, 44.3–67.0] 0 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 15 6 0 Month Data cut-off date: 17 February 2020. No. at risk 84 76 64 56 48 40 35 27 15 9 2 0 CI=confidence interval; ICR=independent central review; laBCC=locally advanced basal cell carcinoma; NE=not evaluable; PFS=progression-free survival. Stratigos AJ, et al. Poster presented at: European Society for Medical Oncology (ESMO) Virtual Congress; September 19–21, 2020:LBA47. Kaplan-Meier Curve for OS by ICR in Patients with laBCC 34 Overall Survival 1.0 0.9 Probability of OS 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 Estimated median OS, months: not reached [95% CI, NE–NE] Estimated 12-month probability of survival: 92.3% [95% CI, 83.6–96.5] 0 0 No. at risk 2 4 6 8 10 12 14 16 18 20 22 24 26 28 51 35 25 15 3 0 0 Month 84 83 80 78 73 72 66 61 Data cut-off date: 17 February 2020. CI=confidence interval; ICR=independent central review; laBCC=locally advanced basal cell carcinoma; NE=not evaluable; OS=overall survival . Stratigos AJ, et al. Poster presented at: European Society for Medical Oncology (ESMO) Virtual Congress; September 19–21, 2020:LBA47. 108 Cemiplimab is the first systemic treatment to show clinical benefit in patients with laBCC and mBCC after HHI therapy. 31% and 21% ORR The safety profile is acceptable for the patient population. Consistent with other PD-1 antibodies and previous reports of cemiplimab in other tumor types Merkel cell carcinoma (MCC) 109 Merkel cell carcinoma (MCC) - Epidemiology Each year ~ 2,500 new cases diagnosed in the EU1 and 1,500 in USA Increasing incidence over the past few decades2 Unclear whether this trend is due to an aging population or increased awareness and diagnosis The highest rates of MCC have been observed in Australia, with an incidence rate of 1.6 per 100,000 persons reported in Queensland3,4 ~80% of MCCs are caused by MCV (Merkel cell polyomavirus) Each year approximately 1 in 3 patients with Merkel Cell Carcinoma will die from their disease 1. IMMOMEC (European Commission). Merkel cell carcinoma. Available at: www.immomec.eu/project/objectives/background/merkel-cell-carcinoma (accessed July 2017); 2. Saini AT, Miles BA. Onco Targets Ther 2015;8:2157–67; 3. Schadendorf D, et al. Eur J Cancer. 2017;71:53–69; 4. Youlden DR, et al. JAMA Dermatol 2014;150:864–72. 37 MCC – Signs, Symptoms and Risk Factors Clinical Presentation Typical presentation –single painless lump on sun-exposed skin1 MCC on the lip FEATURES OF MCC4,5 • Firm, red to purple papule/nodule • Asymptomatic/lack of tenderness • Rapidly Expanding • Mets at an early stage COMMONLY AFFECTED AREAS6 • Head and neck (~50%) • Upper extremities (~16%) • Lower extremities (~30%) • Trunk (<5%) MCC on a cheek RISK FACTORS5 • Immune suppression • Median age ~76 years – Older age • UV exposure • Fair skin . 1. Merkel Cell Carcinoma. National Cancer Institute. Available at: www.cancer.gov/types/skin/patient/merkel-cell-treatment-pdq (accessed December 20163. Image credit: Klaus D. Peter, Gummersbach, Germany. Available at: commons.wikimedia.org/wiki/File:Merkel_cell_cancer.jpg – attribution required for re-use; 4. Nutan FNU et al. Cutis 2014;94:E18–20; 5. Heath M et al. J Am Acad Dermatol 2008;58:375–81; 6. Hitchcock CL et al. Ann Surg 1988;207:201–7. 110 38 Merkel cell carcinoma Where does MCC occur on the body? • MCC primarily occurs on highly sun-exposed skin, but it can occur anywhere on the body, including sunprotected areas such as the buttock or the scalp under hair. Solid circles depict MCC tumors that arose on the skin: 86% of these cases. O Open circles indicate MCCs that presented in lymph nodes without an associated “primary lesion” : 14% of cases 39 Treatment Treatment is generally based on stage of the disease and many issues that are highly variable between patients. It is best to obtain care from a multi-disciplinary team of physicians with significant MCC experience who take into consideration many clinical factors. Major treatments 1) surgical excision of the primary lesion or lymph node, 2) radiation therapy, and 3) systemic therapy including immunotherapy and chemotherapy. 111 Reason for use of immunotherapy in mMCC PD-L1 is expressed in MCC tumor cells and infiltrates of adjacent immune cells1 Dysfunction of MCPyV-specific T cells2 -Levels of CD8 T cells increase with a higher tumor load -Exhausted phenotype (PD-1 +, Tim-3 +) MCPyV-negative tumors have a higher burden on mutations and neoanthigens3 1. Lipson EJ, et al. Cancer Immunol Res. 2013;1(1):54-63; 2. Afanasiev O, et al. Clin Cancer Res. 2014;19(19):5351-60; 3. Goh G, et al. Oncotarget. 2016;7(3):3403-15. JAVELIN MERKEL 200 N=200 (estimated ) A phase II, open-label, multicenter trial to investigate the clinical activity and safety of avelumab (MSB0010718C) in subjects with Merkel cell carcinoma Patients with • Histologicallyproven mMCC • ECOG PS 0–1 Part A: patients have received at least one line of chemotherapy (n=88)2 Part B: patients have not received any prior systemic treatment for mMCC (n=112)1,2 Tumor assessments every 6 weeks (RECIST v1.1; IERC) Part A: PRIMARY ENDPOINT: • Best Overall Response Avelumab 10 mg/kg IV (1-h infusion) every 2 weeks until disease progression, clinical deterioration, unacceptable toxicity or other criterion for withdrawal SECONDARY ENDPOINTS: • DoR, PFS, OS, safety, anti-drug antibodies, PK Part B: PRIMARY ENDPOINT: • Durable response SECONDARY ENDPOINTS: • BOR, DoR, PFS, OS, safety, anti-drug antibodies, PK 42 1. NCT02155647. Available at ClinicalTrials.gov (accessed Oct 2017). 2. Kaufman HL et al. Lancet Oncol 2016;17:1374–85. 112 Part A cohort, mMCC 2L+: design1 n=88 (estimated ) A phase II, open-label, multicenter trial to investigate the clinical activity and safety of avelumab (MSB0010718C) in subjects with Merkel cell carcinoma PRIMARY Part A:ENDPOINT: Avelumab 10 mg/kg IV (1-h infusion) every 2 weeks until disease progression, clinical deterioration, unacceptable toxicity or other criterion for withdrawal Key Inclusion Criteria • Histologically-proven Stage IV MCC • ECOG PS 0–1 • Patients received at least one line of chemotherapy • Durable response* PRIMARY ENDPOINT: KEY SECONDARY ENDPOINTS: • Best Overall Response • BOR**, DoR, PFS, OS, safety, SECONDARY anti-drug antibodies, PK ENDPOINTS: • DoR, PFS, OS, safety, anti-drug antibodies, PK Tumor assessments every 6 weeks (RECIST v1.1; IERC) **Best Overall Response is defined as complete response, partial response, stable disease, or progressive disease, according to RECIST version 1.1 and assessed by an independent review committee. 43 1. NCT02155647. Available at ClinicalTrials.gov (accessed Oct 2017 ). In previously treated patients who received avelumab 52% are still 1 alive at 1 year 100 OS 90 80 OS (%) n=88 Events, n (%) 48 (54.5) 70 Median, months (95% CI) 12.9 (7.5, –) 60 1-year rate, % (95% CI) 52 (41–62) 50 40 Avelumab 30 20 10 0 0 2 4 6 8 10 12 14 16 18 20 22 24 26 3 1 0 Months since initiating treatment Number 88 at risk 81 68 59 50 46 41 27 19 9 6 1. Kaufman HL et al. AACR 2017. Abstract CT079 (presentation). – ≥ 12 month follow-up44 113 Part B cohort, mMCC –Treatment naïve - 1L: design1 n=1121,2 (estimated ) A phase II, open-label, multicenter trial to investigate the clinical activity and safety of avelumab (MSB0010718C) in subjects with Merkel cell carcinoma Key Inclusion Criteria • Histologically-proven Stage IV MCC • ECOG PS 0–1 • Not received any prior systemic treatment for mMCC Avelumab 10 mg/kg IV (1-h infusion) every 2 weeks until disease progression, clinical deterioration, unacceptable toxicity or other criterion for withdrawal Tumor assessments every 6 weeks (RECIST v1.1; IERC) PRIMARY ENDPOINT: • Durable response* KEY SECONDARY ENDPOINTS: • BOR, DoR, PFS, OS, safety, anti-drug antibodies, PK Estimated primary completion date: September 2019 * By Kaplan Meier Method *Durable response is defined as proportion of patients of the total ITT population with objective response (CR or PR) according to RECIST v1.1, with a duration of at least 6 months.* * 45 1. NCT02155647. Available at ClinicalTrials.gov (accessed Oct 2017). 2. Kaufman HL et al. Lancet Oncol 2016;17:1374–85. Avelumab in 1L mMCC: High ORR 62.1% and >80% on-going for at least 6 months CR in 4 patients Preliminary Efficacy Data – Treatment naïve – 1L1 1. D'Angelo SP, et al. Efficacy and Safety of First-lineAvelumab Treatment in Patients With Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysisof a Clinical Trial. JAMA Oncol 2018 Sep 1;4(9):e180077; 114 46 In this multicenter phase II trial (Cancer Immunotherapy Trials Network-09/Keynote- 017), 50 adults na¨ıve to systemic therapy for aMCC received pembrolizumab (2 mg/kg every 3 weeks) for up to 2 years. Radiographic responses were assessed centrally per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. J Clin Oncol 37:693-702. 2019 ORR to pembrolizumab was 56% (complete response [24%] plus partial response [32%]; 95% CI, 41.3% to 70.0%), with ORRs of 59% in virus-positive and 53% in virus-negative tumors. Median follow-up time was 14.9 months (range, 0.4 to 36.4+ months). Among 28 responders, median response duration was not reached (range, 5.9 to 34.5+ months). The 24-month PFS rate was 48.3%, and median PFS time was 16.8 months (95% CI, 4.6 months to not estimable). The 24-month OS rate was 68.7%, and median OS time was not reached. Although tumor viral status did not correlate with ORR, PFS, or OS, there was a trend toward improved PFS and OS in patients with programmed death ligand-1–positive tumors. Grade 3 or greater treatment-related adverse events occurred in 14 (28%) of 50 patients and led to treatment discontinuation in seven (14%) of 50 patients, including one treatment-related death. J Clin Oncol 37:693-702. 2019 115 J Clin Oncol 37:693-702. 2019 In patients with aMCC receiving first-line anti– programmed cell death-1 therapy - Pembrolizumab demonstrated durable tumor control, a generally manageable safety profile, and favorable OS compared with historical data from patients treated with first-line chemotherapy. J Clin Oncol 37:693-702. 2019 116 Nivolumab trial: CheckMate358 (NCT02488759) Non-comparative, two-cohort, single-arm, open-label, Phase I/II study of nivolumab (BMS-936558) in subjects with virus-positive and virus-negative solid tumors N=500 Tumor types: MCC, gastric or GEJ carcinoma, nasopharyngeal carcinoma, SCC of cervix, vagina or vulva, SCCHN NEOADJUVANT COHORT Nivolumab METASTATIC COHORT Nivolumab PRIMARY ENDPOINT Safety and tolerability, ORR in metastatic patients, surgery delay rate SECONDARY ENDPOINTS Change in immune cells of viral-specific T-cells; change in immune activation/inhibitory molecules of viral-specific T-cells; PFS; OS • NCT02488759. Available at: ClinicalTrials.gov (accessed December 2016). 51 Nivolumab in 1L advanced MCC - Efficacy RESPONSE* (n=15) ORR, % (95% CI) 73 (45–92) CR, n (%) PR, n (%) 7 (47) 4 (27) *By RECIST v1.1, investigator assessed 1. Topalian SL et al. AACR 2017. Abstract CT074 (presentation). 117 52 Nivolumab* in 2L+ - Efficacy RESPONSE* (n=10) ORR, % (95% CI) 50 (19–81) CR, n (%) PR, n (%) 10 (1) 40 (4) *By RECIST v1.1, investigator assessed 1. Topalian SL et al. AACR 2017. Abstract CT074 (presentation). 53 Immunotherapy Phase II of the JAVELIN Merkel 200 trial studied Avelumab in patients with metastatic MCC either as 1st line therapy1 or in chemotherapy-refractory MCC2-3. In patients with no prior systemic therapy, after a median follow-up of 5.1 months (range 0.3-11.3 months), the overall response rate was 62.1%, and 83% of patients had a duration of response of at least 6 months1. In patients treated with avelumab after progression on chemotherapy, the overall response rate was 33.0% after a minimum follow up of 12 months. At the time of data cut-off, 72.4% of responses were ongoing3 A different phase II trial studied patients with advanced MCC treated with pembrolizumab4; - after a median follow up of 33 weeks (range 7-53 weeks) the overall response rate was 56%, with a response duration ranging from 2.2-9.7 months. The ongoing CHECKMATE 358 phase I/II trial is studying nivolumab in patients with resectable MCC5. In pts treated with nivolumab prior to surgery, 80% had tumour regression and 65% had a major pathologic response including 8 CR. 1.D'Angelo SP, et al. Efficacy and Safety of First-lineAvelumab Treatment in Patients With Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysisof a Clinical Trial. JAMA Oncol 2018 Sep 1;4(9):e180077; 2.Kaufman HL, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial.Lancet Oncol 2016 Oct;17(10):1374-1385; 3. Kaufman HL, et al. Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after >/=1 year of follow-up: JAVELIN Merkel200, a phase 2 clinical trial. J Immunother Cancer 2018 Jan 19;6(1):x. 4.Nghiem PT, et al. PD-1 Blockade withPembrolizumab in Advanced Merkel-Cell Carcinoma. N Engl J Med 2016 Jun 30;374(26):2542-2552. 5.Topalian SL, Bhatia S, Kudchadkar RR, Amin A, Sharfman WH, Lebbe C, et al. Nivolumab (Nivo) as neoadjuvanttherapy in patients with resectable Merkel cell carcinoma (MCC) in CheckMate 358. JCO 2018;36(15):9505. 118 Anti PD-1/PD-L1 in advanced MCC ORR 1st line 56-73% 2nd line 33-50% PFS 1st line 17 mo (median) 2nd line 3 mo (median) OS 1st line median not reached 2nd line 13 mo (median) 119 Summary MCC NMSC - the most common cancer Incidence is rising Numerous mutations in UV-induced cancer Surgery is a standard therapy for non-complicated cases Limited role of radiotherapy despite radiosensitivity in MCC anti-PD-1/PD-L1 should be applied as first-line treatment ChT should be postponed to 2nd line Previous ChT impairs outcome of anti-PD-1/PD-L1 Summary NMSC There is no clear benefit of chemotherapy Targeted therapy in BCC patched / SMOi is effective (RR 58%, CR 20-30%) Imunoterapies (PD-1 and PD-L1 antibodies) are effective in MCC and SCC, they also promise a great deal of potential for BCC 120 Our experience & interesting cases Maša Sever, Janja Ocvirk Vismodegib in Routine Clinical Practice in Slovenia 121 Although basal cell carcinoma (BCC) is the most common malignancy among Caucasians, the incidence of advanced forms is relatively rare and therefore there is little data in the literature on treatment of locally advanced BCC (laBCC) or metastatic BCC (mBCC) by systemic therapy. Vismodegib is a Hh signaling pathway inhibitor and was approved by the European Medicines Agency for the treatment of adults with mBCC, or with laBCC inappropriate for surgery or radiotherapy. A retrospective analysis was conducted to provide vismodegib longterm efficacy and safety data in a real-world setting in Slovenia. Methods Evaluation of efficacy and safety of vismodegib (V) was done in a retrospective analysis of patients (pts) with laBCC or multiple BCC and pts with Goltz-Gorlin Syndrom (G-G Syn) in routine clinical practice. Baseline characteristics, efficacy data and treatment-related adverse events (AEs) were collected from pts who were treated with V from November 2012 to January 2021. Efficacy was assessed by objective response rate - ORR (CR + PR), disease control rate - DCR (CR + PR + SD) and duration of vismodegib treatment – DoT. Reasons for treatment discontinuation were analyzed. 122 Baseline characteristics During the 100-month period, 46 pts were diagnosed with laBCC (26 pts), multiple BCC (13 pts) or G-G Syn (7 pts), all inappropriate for surgery or radiotherapy. Baseline characteristics: median age was 72.8 years in laBCC + multiple BCC pts group and 47.4 years in the G-G Syn pts group (Fig.1). Fifty-six percent of pts in laBCC +multiple BCC group were females; the majority (67%) of pts were previously treated by surgery and/or radiotherapy; 51% of pts had one lesion with predominant localization in the central face area (eyes, nose, lips, or ears in 76% of pts), 18% had 2-3 lesions, and 31% more than 3 lesions. Fifty-seven percent of pts in G-G Syn group were males; 86% of pts were previously treated with surgery and/or radiotherapy. Results Figure 1. Age distribution at vismodegib treatment initiation Patients with Goltz-Gorlin Syndrom (N=7) 16 14 12 10 8 6 4 2 0 4 Number of patients Number of patients Patients with laBCC and multiple BCC (N=39) 3 2 1 0 30-39 40-49 50-59 60-69 70-79 80-89 90- 20-29 30-39 Age distribution Efficacy results 40-49 50-59 60-69 Age distribution • At the time of analysis in laBCC or multiple BCC group treatment has been interrupted during the treatment course in 23% of pts [in 8 out of 9 pts due to adverse events (AEs)], 31% of pts are still on treatment. In G-G Syn group treatment has been interrupted in 57% of pts (in most cases due to adverse events), 43% of pts are still on treatment. Efficacy data are presented in Table 1. Table 1. Response to vismodegib treatment laBCC + multiple BCC group (N=39) Efficacy variable Investigator-assessed objective response rate (CR+PR); n, % Investigator-assessed disease control rate (CR+PR+SD); n, % Duration of treatment (months); median (range) 123 G-G Syn group (N=7) 31 (80%) 6 (86%) 37 (95%) 7 (100%) 9.9 (1.5-43.1) 19.5 (3.6-94.1) Safety results Serious adverse events were reported in 6 out of 46 patients (13.0%): two cases of squamous cell cancer, one case of angiosarcoma, melanoma, cholangiocarcinoma and intracerebral hemorrhage each, while one patient died due to other reasons than cancer. AEs of any grade were reported in 82% of pts in laBCC or multiple BCC group and 71% in G-G Syn group. The majority of AEs in laBCC or multiple BCC group were grade 1 or 2 (96%) and only 4% of AEs were grade 3: muscle cramps in 3 pts, respiratory infection, vomiting and anemia in 1 patient each. The majority of AEs in G-G Syn group were also grade 1 or 2 (87%), while 13% of AEs were grade 3: muscle cramps in 2 pts, weight loss and diarrhea in 1 patient each. No grade 4 or 5 vismodegib related AEs were reported. Conclusion Vismodegib has shown meaningful long-term efficacy with manageable safety profile in pts with laBCC or multiple BCC as well as in pts with G-G Syn in real-world setting. 124 Clinical cases BCC 125 19. 12. 2013 Case from OIL 31. 7. 2014 23. 9. 2013 Quick response to high-dose treatment Side effects: alopecia gr. 2 after one year of treatment, increased CPK gr.1, muscle cramps gr.1 11. 2012 Case8.from OIL 11 16. 10. 2014 Patient with Gorlin syndrome (multiple BCC) Side effects: alopecia gr.1 weight loss gr.2 increased CPK gr.1-3 12 126 Side effects: alopecia gr. 2, muscle spasms gr.2, change in taste gr.1 Case from OIL 21.11.2013 25.9.2014 13 Man, 87 years old History: 2001 – Multipl epiteliomas left leg 2001 – Malignant melanoma left leg/excision 2011 – BCC/SCC left leg/ excision, RT → ulcus BCC right leg → Not suitable for RT or excision Others: 2003 – Nefrectomy 2006 – Deep vein thrombosis 2012 – Deep vein thrombosis 2012 – hypertension therapy: varfarin and trandolapril since 2012 14 127 January 2018 July 2019 March 2019 128 August 2020 October 2020 January 2020 129 May 2020 La BCC After 3 m vismodegib BCC – 2 lines of treatment After 6 w of immunotherapy Progres on vismodegib, Start of immunotherapy After 3 m of immunotherapy October 2018 MCC Case October 2018 Male 68-year-old, presented with bleeding tumor on right thigh, invasions in inguinal lymph nodes, obturator lymph nodes, right iliac lymph nodes Biopsy - MCC TMB decision: immunotherapy- avelumab 130 before starting immunotherapy marked pain occurred bleeding from the tumor the surgeons decided for resection of a bleeding tumor on the thigh complication of treatment for sepsis, wound dehiscence he was recovering from complications of surgical treatment in January 2019 referred to medical oncologist the patient also has rheumatoid arthritis on methotrexate therapy highly motivated for immunotherapy treatment he began with immunotherapy (Avalumab) in January 2019 CT scan in April 2019 - PRCT January 2019 scan in July 2019 – further response CT scan in october 2019 - borderline enlarged lymph node left inguinal, no other evident residum of the disease Stop immunotherapy due to AE in March 2020 131 16.10.2018 27.8.2019 SCC 132 11.10.2020 Start treatment with cemiplimab 2.11.2020 After 1 application 22.2.2021 After 2 appl and post COVID pneumonia Thank you for your attention 133 7.6.2021 CR confirmed with biopsy NEXT-GENERATION SEQUENCING Assist. Prof. Tanja Mesti, MD., PhD 2nd Summer school 7 September 2021 Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group ◦ ESMO Scale for Clinical Actionability of molecular Targets (ESCAT) ◦ Advanced lung carcinoma ◦ Metastatic prostate cancer ◦ Metastatic ovarian cancer ◦ Metastatic cholangiocarcinoma *cancer types without clear standard-of-care options, such as carcinoma of unknown primary and other rare tumors Annals of Oncology 2020 311491-1505DOI: (10.1016/j.annonc.2020.07.014) 134 CONCLUSION ◦ ESMO Guidelines ◦ NGS consilium - subgroup of patients with advanced cancer and no standard therapeutic options. 135 Systemic treatment of gastric carcinoma Marko Boc, MD Department for Medical Oncology Institute of Oncology Ljubljana Ljubljana, 08.09.2021 NEOADJUVANT AND PERI-OPERATIVE CHEMOTHERAPY ◆ ◆ ◆ ◆ Downstage the tumour Increase R0 resection rate Treat micrometastatic disease Improve overall survival Neoadjuvant and perioperative chemotherapy is more commonly used in non-Asian countries where tumours are frequently locally advanced and require downstaging prior to successful resection 136 NEOADJUVANT AND PERI-OPERATIVE CHEMOTHERAPY 5 year OS 45%* 5 year OS 38%* 5 year OS 36%* 5 year OS 23%* 2002 2 cycles neoadjuvant CF (OE02) 2011 3+3 cycles CF (ACCORD) 2006 3+3 cycles ECF (MAGIC) *chemotherapy patients; CF, cisplatin + 5-fluourouracil; ECF epirubicin + CF; FLOT. 5-fluororuraci, leucovorin, oxaliplatin, docetaxel ADJUVANT CHEMOTHERAPY FOR NON-ASIAN PATIENTS 2017 4+4 cycles FLOT (FLOT4) 1. OE02 Trial Group, Lancet 2002. 2. Cunningham D, et al. N Engl J Med 2006. 3. Ychou M, et al. J Clin Oncol. 2011. 4. Al-Batran S, et al. Lancet 2019. ADJUVANT CHEMOTHERAPY Although non-Asian adjuvant gastric cancer trials have not shown a survival benefit, this metanalysis can be used to justify use of adjuvant chemotherapy in non-Asian patients Paolettiet al, JAMA. 2010 May 5;303(17):1729-37. 137 mOS: 8-11m mPFS: 4.8-6.2m 138 2nd Summer School in medical oncology – Precision oncology – Are we there yet CLINICAL CASE GASTRIC CANCER Ana Erman, dr. med. Mentor: doc. dr. Tanja Mesti, dr. med. Ljubljana, 8.9.2021 SUMMARY November 2015 March 2016 May 2016 July 2017 October 2018 April 2018 January 2021 Peritoneal carcinomatosis Gastric and esophageal adenocarcinoma T4NxM0 Neoadjuvant CRT Capecitabine and Trasuzumab Reinduction of paclitaxel PFS 14 months PFS 6 months st 1 line systemic therapy: Capecitabine, Paclitaxel and ramucirumab oxaliplatin and PFS 16 months Immunotherapy - Pembrolizumab trastuzumab PFS 14 months 139 SUMMARY • HER2, MSI and NTRK testing • If sufficient tissue: NGS • Pembrolizumab for MSI-H/dMMR tumors or TMB-high (>10 mutations/megabase) tumors 140 Systemic treatment of the Biliary tract cancer (BTC)– where we stand Summer School, 8th September 2021 assist.prof. Martina Reberšek, MD, PhD Current recommendations for systemic treatment of metastatic disease Systemic chemotherapy: - 1st line: gemcitabine + cisplatin (PS ECOG 0-1), gemcitabine mono (PS ECOG 2) - 2nd line: mFOLFOX or new standard option Nal-IRI+5FU/LV Immunotherapy: - 1st line: MSI-H/dMMR → pembrolizumab - 2nd line: nivolumab, MSI-H/dMMR/TMB-H → pembrolizumab Targeted therapy - 1st line: positive NTRK fusion gene →larotrectinib, entrectinib - 2nd line: →posi:ve NTRK fusion gene → larotrectinib, entrectinib → mt BRAF V600 → dabrafenib+trametinib → mt IDH1 → ivosidenib →FGFR2 fusions or rearrangements → pemigatinib 141 How to approach GEP NET & NEC MARIJA IGNJATOVIĆ. 2ND SUM M ER SCHO O L IN M EDICA L O NCO LO G Y 08.09.2021 NEUROENDOCRINE NEOPLASMS (NENs)  Heterogeneous group of malignancies ► arise from neuroendocrine cells ► release of catecholamines and hormones  Rare malignancies  Less then 0.5% of all malignancies, only 1%-2% of gastrointestinal malignancies  Growing incidence, especially of localized and regional NENs  EUR 1.33 – 2.33/100.000; USA: 3.56/100.00  men > women, men have adverse outcome  Sporadic/hereditary  Younger then < 40 with gastrinoma  Multiple neuroendocrine neoplasia (MEN)  Family history of NENs 142 CLASSIFICATION OF GASTROENTEROPANCREATIC NEOPLASMS (GEP-NENs) FIRST MORPHOLOGY Well-differentiated & poorly-differentiated THEN KI67 Grade1/2/3 WHO 2019 MORPHOLOGY WELL DIFFERENTIATED POORLY DIFFERENTIATED GRADE MITOTIC INDEX (2 mm²) KI 67 (%) G1 <2 <3 G2 2-20 3-30 G3 >20 >20 G3 >20 >20 Neuroendocrine tumors (NET) Neuroendocrine carcinomas (NEC) MiNEN GASTROENTEROPANCREATIC NEOPLASMS (GEP-NENs) 143 GEP-NET ► 80% - 90% GEP-NEC ► 10% - 20% YOUR PATIENTS MIGHT BE (A)SYMPTOMATIC HYSTOPATHOLOGY • Morphology • Grade • IHC DIAGNOSIS 144 STAGING PET CT with GALLIUM 68/SRS FDE-PETCT MRI CT MANAGEMENT OF PATIENTS WITH LOCAL & LOCOREGIONAL GEP NETs 145 SURGERY MANAGEMENT OF PATIENTS WITH LOCAL & LOCOREGIONAL GEP NECs MANAGEMENT OF PATIENTS WITH METASTATIC GEP NET 146 SURGERY SYSTEMIC TREATMENT CONTROL OF HORMONAL OVERPRODUCTION Systemic treatment and mGEP-NETs CONTROL OF TUMOUR GROWTH MENAGMENT OF PATIENTS WITH mGEP NEC 147 SYSTEMIC TREATMENT How to approach NET/NEC Our experience – clinical case Katja Leskovšek, dr. med. prof. dr. Janja Ocvirk, dr. med. Institute of Oncology Ljubljana, 08.09.2021 D.N., female, 54y  On presentation, Nov 2005  6 months of diarrhoea, loss of weight (10 kg).  Family history: positive.  Past medical history: asthma (2003), discus hernia L4-5 op. (1999).  PS ECOG 1.  Therapy: Symbicort.  Alergies: ketoprofen.  US: 3 liver metastases Core needle biopsy: NEC metastases; origo ignota.  Laparotomy (Sept 2005): tumor of distal pancreas, invading spleen  Distal pancreatectomy and splenectomy, resection of IV. and V. liver segment, cholecystectomy. 148 D.N., female, 54y  Histopathology report: NEC of pancreas, invading retroperitoneal fat, hilum of spleen, vascular, peri- and intraneural invasion, lymphangiosis carcinomatosa; lymph node status 3/5.  R1 resection.  Somatostatin: 10 %.  Partial liver resection: metastasis of endocrine carcinoma. Discussion  NEC of pancreas, liver metastasis, R1.  On treatment for 15 years.  Systemic therapy:   I. line ChT: etoposide + cisplatin;  II. line ChT: 5-FU + dacarbazine + epidoxorubicin;  3 x partial liver resection/metastasectomy (12‘, 13‘, 13‘);  III. line: lanreotide;  IV. line: lanreotide + everolimus;  V. line: octreotide + sunitinib;  6 x TACE (19‘, 20‘, 21‘); PRRT ? 149 HCC – LOTS HAS BEEN GOING ON Janja Ocvirk INCIDENCA 150 HCC The fourth most common cause of cancer-related death worldwide1 HCC accounts for >80% of primary liver cancers worldwide1 Chronic HBV and HCV infection are the most important causes of HCC and account for 80% of HCC cases globally1 It is estimated that 72% of cases occur in Asia (more than 50% in China)2 Staging of HCC is important to determine outcome and planning of optimal therapy. While there are a number staging systems used, the BCLC is currently commonly used to compare clinical outcomes:3 ◦ 1. 2. 3. BCLC, Barcelona Clinic Liver Cancer; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; TACE, transarterial chemoembolisation Yang JD, et al. Nat Rev Gastroenterol Hepatol. 2019;16:589-604 Singal AG, et al. J Hepatol. 2020;72:250-61 Bruix J, et al. Nat Rev Gastroenterol Hepatol. 2019;16:617-30 BCLC Staging System 151 3 HCC Early and intermediate HCC BCLC staging Survival rate with current therapy Standard of care treatment Stage 0-A >5 years Ablation, resection, transplantation Stage B >2.5 years Chemoembolisation (TACE) Stage C >1 year Systemic therapy Stage D 3 months Best supportive care Advanced HCC ◦ 1. 2. 3. BCLC, Barcelona Clinic Liver Cancer; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; TACE, transarterial chemoembolisation Yang JD, et al. Nat Rev Gastroenterol Hepatol. 2019;16:589-604 Singal AG, et al. J Hepatol. 2020;72:250-61 Bruix J, et al. Nat Rev Gastroenterol Hepatol. 2019;16:617-30 1L FOR ADVANCED HCC PATIENTS 152 5 Phase 3 SHARP trial of sorafenib vs placebo: study design1 Eligibility •Advanced HCC* •Child–Pugh A •ECOG PS 0–2 •No prior systemic therapy Stratification •Geographic region •ECOG PS (0 vs 1-2) •MVI/EHS (present/absent) N=602 R A N D O M I Z E Sorafenib 400 mg BID (n=299) Placebo (n=303) 1:1 Primary endpoints •OS •TTSP† Secondary endpoints •TTP •DCR •Safety‡ Note: same study design was used for SHARP-AP2 *Not eligible for, or had disease progression after surgical or locoregional therapies. †Assessed by the Functional Assessment of Cancer Therapy-Hepatobiliary Symptom Index-8 (FSHI-8). ‡Assessed using version 3.0 of the USA National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE). BID, twice daily; DCR, disease control rate; EHS, extrahepatic spread; MVI , macroscopic vascular invasion; OS, overall survival; TTP, time to treatment progression; TTSP, time to symptomatic progression. 1. Llovet JM et al. N Engl J Med. 2008;359(4):378-390; 2. Cheng A et al. Lancet Oncol 2009;10:25–34. SHARP: Baseline Patient Characteristics BCLC stage (stage B: 18.1% vs. 16.8%; stage C: 81.6% vs. 83.2%; stage D: <1% vs. 0%) in sorafenib and placebo respective Llovet JM, et al. N Engl J Med. 2008;359:378-390. 153 SHARP - efficacy data Efficacy parameter Median OS, months (95% CI) Median TTP, months (95% CI) Sorafenib (n=299) Placebo (n=303) P-value 10.7 (9.4-13.3) 7.9 (6.8-9.1) 0.00058 0.69 (0.55-0.87) 5.5 (4.1-6.9) 2.8 (2.7-3.9) 0.000007 0.58 (0.45-0.74) 1. Llovet JM et al. N Engl J Med. 2008;359(4):378-390; 2. HR (95% CI) 9 SHARP: sorafenib is generally well tolerated in advanced HCC Incidence by grade (%) (N=297) Adverse event* Any grade Grade 3/4 Diarrhoea 39 8† Fatigue 22 4 HFSR 21 8† Rash/desquamation 16 1† Anorexia 14 <1† Abdominal pain 8 2† Liver dysfunction <1 <1† Nausea 11 <1† Vomiting 5 1† Weight loss 9 2† Hypertension 5 2† *Defined by NCI CTC (version 3.0) that occurred in at least 5% of patients; †No grade 4 events reported. HFSR, hand–foot skin reaction. Llovet JM et al. N Engl J Med 2008;359:378–90; EASL–EORTC Clinical Practice: Management of hepatocellular carcinoma. J Hepatol 2012;56:908–43; Verslype C et al. ESMO guidelines. Ann Oncol 2012;23(Suppl 7):vii41–8. 154 Data from SHARP and real-world practice support use of sorafenib in intermediate HCC SHARP1 BCLC-B subgroup •Increased OS and TTP with sorafenib (n=54) vs placebo (n=51) – Median OS: 14.5 vs 11.4 months (HR: 0.72; 95% CI: 0.38–1.38) – Median TTP: 6.9 vs 4.4 months (HR: 0.47; 95% CI: 0.23–0.96) SHARP1 previous TACE subgroup •Increased OS and TTP with sorafenib (n=86) vs placebo (n=90) – Median OS: 11.9 vs 9.9 months (HR: 0.75; 95% CI: 0.49–1.14) – Median TTP: 5.8 vs 4.0 months (HR: 0.57; 95% CI: 0.36–0.91) SOFIA2 •Good efficacy demonstrated in BCLC-B HCC – Longer survival in BCLC-B vs BCLC-C patients: 20.6 vs 8.4 months INSIGHT3 •Good efficacy demonstrated in BCLC-B HCC – Longer survival in BCLC-B vs BCLC-C patients: 19.6 vs 14.5 months GIDEON interim analysis4 •Similar safety profile for sorafenib across BCLC stages BCLC, Barcelona Clinic Liver Cancer; HCC, hepatocellular carcinoma; HR, hazard ratio; OS, overall survival; TTP, time to progression 1. Bruix J et al. J Hepatol. 2012;57:821–9; 2. Iavarone M et al. Hepatology 2011;54:2055–63; 3. Ganten TM et al. EMSO 2012;poster 77; 4. Lencioni R et al. Eur J Cancer 2011;47 (Suppl 1):abstract 6500 155 Lenvatinib - REFLECT REFLECT (NCT01761266): phase 3, international, multicentre, open-label, randomised study in 954 patients with hepatocellular carcinoma Non inferiority assessment of lenvatinib vs. sorafenib for OS Primary endpoint: OS Secondary endpoints: PFS, ORR (mRECIST and RECIST v1.1) Population enrolled: BCLC stage B: 20%; stage C: 80% BCLC, Barcelona Clinic Liver Cancer; CI, confidence interval; mRECIST, modified RECIST; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; RECIST, Response Evaluation Criteria in Solid Tumours Sources: lenvatinib summary of product characteristics dated June 2020; lenvatinib US prescribing information dated February 2020 Efficacy parameters Overall survival Number of deaths (%) Median OS in months (95% CI) Hazard ratio (95% CI) Progression-free survival (mRECIST) Number of events (%) Median PFS in months (95% CI) Hazard ratio (95% CI) and P-value Objective response rate (mRECIST) Objective response rate Complete responses, n (%) Partial responses, n (%) 95% CI P-value Progression-free survival (RECIST 1.1) Number of events (%) Median PFS in months (95% CI) Hazard ratio (95% CI) Objective response rate (RECIST 1.1) Objective response rate Complete responses, n (%) Partial responses, n (%) 95% CI Lenvatinib Sorafenib N=478 N=476 13 351 (73) 350 (74) 13.6 (12.1-14.9) 12.3 (10.4-13.9) 0.92 (0.79-1.06) 311 (65) 323 (68) 7.3 (5.6-7.5) 3.6 (3.6-3.7) 0.64 (0.55-0.75); <0.001 41% 10 (2.1) 184 (38.5) (36-45) 12% 4 (0.8) 55 (11.6) (10-16) <0.001 307 (64) 7.3 (5.6-7.5) 320 (67) 3.6 (3.6-3.9) 0.65 (0.56-0.77) 19% 2 (0.4) 88 (18.4) (15-22) 7% 1 (0.2) 30 (6.3) (4-9) ◦ BCLC, Barcelona Clinic Liver Cancer; CI, confidence interval; mRECIST, modified RECIST; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; RECIST, Response Evaluation Criteria in Solid Tumours ◦ Sources: lenvatinib summary of product characteristics dated June 2020; lenvatinib US prescribing information dated February 2020 14 156 Sorafenib and lenvatinib safety data in HCC patients Most common adverse reactions (≥20%) sorafenib-treated patients in SHARP trial Diarrhoea – fatigue – hand-foot skin reaction – weight loss – anorexia – nausea – abdominal pain lenvatinib-treated patients in REFLECT trial Hypertension – fatigue – diarrhoea – decreased appetite – arthralgia/myalgia – decreased weight – abdominal pain – palmar-plantar erythrodysesthesia syndrome – proteinuria – dysphonia – haemorrhagic events – hypothyroidism – nausea ◦ Sources: sorafenib SmPC; lenvatinib SmPC 15 IMbrave150 clinical trial Key eligibility criteria • Locally advanced metastatic or unresectable HCC • ECOG PS 0-1 • No prior systemic therapy • No bleeding or high risk of bleeding (n=501) Stratification criteria • Region: Asia (excluding Japan) or rest of world • ECOG PS 0 or 1 • Presence or absence of macrovascular invasion or extrahepatic spread atezolizumab 1200 mg IV q3w + bevacizumab 15 mg/kg IV q3w R (Open label) 2:1 sorafenib Until loss of clinical benefit or unacceptab le toxicity Survival followup 400 mg BID • Baseline AFP <400 or ≥400 ng/mL Co-primary endpoints • OS • IRF-assessed PFS per RECIST 1.1 Secondary endpoints include • IRF-assessed ORR per RECIST 1.1 and HCC mRECIST • PROs AFP, alpha-fetoprotein; BID, twice a day; ECOG PS; Eastern Cooperative Oncology Group performance status; HCC; hepatocellular carcinoma; IRF, independent review facility; IV, intravenous; mRECIST, modified RECIST; ORR, objective response rate; OS, overall survival; PD-L1, programmed death-ligand 1; PFS, progression-free survival; PRO, patient-reported outcome; q3w, every 3 weeks; R, randomization; RECIST, Response Evaluation Criteria In Solid Tumours; VEGF, vascular endothelial growth factor Finn RS, et al. N Engl J Med. 2020;382:1894-905 157 16 IMbrave150 clinical trial Efficacy results: primary endpoints atezolizumab + bevacizumab (n=336) sorafenib (n=165) NE 13.2 (10.4–NE) Median OS (95% CI), months 0.58 (0.42–0.79) OS, HR (95% CI) <0.001 P-value 6.8 (5.7–8.3) Median PFS (95% CI) per IRF RECIST v1.1, months 4.3 (4.0–5.6) 0.59 (0.47–0.76) PFS, HR (95% CI) <0.001 P-value ◦ Finn RS, et al. N Engl J Med. 2020;382:1894-905 17 IMbrave150 clinical trial Efficacy results: secondary endpoints Confirmed ORR per IRF RECIST v1.1, % (95% CI) atezolizumab + bevacizumab (n=326) sorafenib (n=159) 27.3 (22.5–32.5) 11.9 (7.4–18.0) <0.001 P-value Confirmed ORR per HCC specific mRECIST, % (95% CI) atezolizumab + bevacizumab (n=325) sorafenib (n=158) 33.2 (28.1–38.6) 13.3 (8.4–19.6) P-value <0.001 ◦ Finn RS, et al. N Engl J Med. 2020;382:1894-905 18 158 IMbrave150 clinical trial - safety results atezolizumab + bevacizumab (n=329) sorafenib (n=156) Patients with an AE from any cause 323 (98.2) 154 (98.7) Grade 3-4 AEs (numbers represents the highest grades assigned) 186 (56.5) 86 (55.1) 15* (4.6) 9** (5.8) Serious adverse event 125 (38.0) 48 (30.8) AEs leading to withdrawal from any trial drug 51 (15.5) 16 (10.3) AEs leading to dose modification or interruption of any trial drug 163 (49.5) 95 (60.9) 163 (49.5) 64 (41.0) – 58 (37.2) Variables, n (%) Grade 5 AEs Dose interruption of any trial treatment Dose modification of sorafenib *Grade 5 events in the atezolizumab–bevacizumab group: gastrointestinal haemorrhage (in 3 patients), pneumonia (in 2 patients), empyema, gastric ulcer perforation, abnormal hepatic function, liver injury, multiple-organ dysfunction syndrome, oesophageal varices haemorrhage, subarachnoid haemorrhage, respiratory distress, sepsis, and cardiac arrest (in 1 patient each) ◦ ◦ **Grade 5 events in the sorafenib group: death (in 2 patients), hepatic cirrhosis (in 2 patients), cardiac arrest, cardiac failure, general physical health deterioration, hepatitis E, and peritoneal haemorrhage (in 1 patient each) AEs, adverse events Finn RS, et al. N Engl J Med. 2020;382:1894-905 19 IMbrave150 clinical trial - conclusion ◦ IMbrave150 demonstrated a statistically significant improvement in OS and PFS with atezolizumab + bevacizumab versus sorafenib in the first-line setting in patients with advanced HCC ◦ Times to response were similar in the combination and sorafenib arms ◦ Response rates were significantly higher in the combination arm ◦ The trial was conducted in a patient population that had preserved liver function (Child–Pugh class A) and a decreased risk of variceal bleeding. The safety of the combination in a broader population warrants further study Finn RS, et al. N Engl J Med. 2020;382:1894-905 20 159 Atezolizumab + bevacizumab vs sorafenib for unresectable hepatocellular carcinoma (HCC): Results from older adults enrolled in IMbrave150 Overall survival Key results Age <65 years Median OS, mo HR (95%CI) Sorafenib (n=74) NE 11.4 Median OS, mo 0.59 (0.38, 0.91) 80 80 60 60 40 20 0 0 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 Time, months No. at Atezo risk 175172169163156147140127107 81 55 42 30 16 8 4 2 NE + bev 2 1 NE NE Atezo + bev (n=161) Sorafenib (n=91) NE 14.9 0.58 (0.36, 0.92) 40 20 SOR 74 69 62 56 54 51 44 36 33 25 20 16 11 7 HR (95%CI) 100 OS, % OS, % 100 Age ≥65 years Atezo + bev (n=175) 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 Time, months 161157151149146141135128115 84 63 45 34 24 12 7 1 NE 91 88 81 76 73 67 61 58 53 35 25 17 13 9 1 NE 5 2 Li D, et al. Ann Oncol 2020;31(suppl):abstr O-8 Atezolizumab + bevacizumab vs sorafenib for unresectable hepatocellular carcinoma (HCC): Results from older adults enrolled in IMbrave150 AEs occurring in ≥15% of patients treated with atezolizumab + bevacizumab, n (%) <65 years (n=171) ≥65 years (n=158) Hypertension 47 (27) 51 (32) Fatigue 24 (14) 43 (27) Diarrhoea 28 (16) 34 (22) Appetite decreased 26 (15) 32 (20) Pyrexia 29 (17) 30 (19) Pruritus 35 (20) 29 (18) Proteinuria 39 (23) 27 (17) AST increased 39 (23) 25 (16) Conclusions ◦ In older patients (≥65 years) with unresectable HCC, atezolizumab + bevacizumab demonstrated clinically meaningful benefits with no significant additional toxicities Li D, et al. Ann Oncol 2020;31(suppl):abstr O-8 160 CheckMate 459: long-term efficacy outcomes with nivolumab versus sorafenib as firstline treatment in patients with advanced hepatocellular carcinoma – Sangro B, et al Study objective ◦ To evaluate the long-term efficacy and safety of nivolumab as a 1L treatment for patients with advanced HCC Key patient inclusion criteria Nivolumab 240 mg iv q2w (n=371) • Advanced HCC • Ineligible for surgery and/or for loco-regional therapy or PD after surgery and/or loco-regional therapy PD/ toxicity Stratification • Aetiology (HCV vs. non-HCV) • Vascular invasion and/or extrahepatic spread (present vs. absent) • Geography (Asia vs. non-Asia) R 1:1 • Child-Pugh class A • Systemic therapy naïve Sorafenib 400 mg po bid (n=372) • ECOG PS 0–1 PD/ toxicity (n=743) SECONDARY ENDPOINTS • ORR, PFS, efficacy by PD-L1 status, safety PRIMARY ENDPOINT • OS Sangro B, et al. Ann Oncol 2020;31(suppl):abstr LBA-3 This talk was presented at the 22nd ESMO WCGC on 1 July 2020 at 18:20 CheckMate 459: long-term efficacy outcomes with nivolumab versus sorafenib as firstline treatment in patients with advanced hepatocellular carcinoma – Sangro B, et al Overall survival Nivolumab (n=371) Key results Primary analysis: June 2019 database lock 100 Median OS, months (95%CI) 16.4 (13.9, 18.4) HR (95%CI); p-value 80 12-mo rate 60 Median OS, months (95%CI) 37% 16.4 (14.0, 18.5) HR (95%CI); p-value 24-mo rate 55% 14.7 (11.9, 17.2) 0.85 (0.72, 1.02); 0.0752 Long-term follow-up analysis: April 2020 database lock 60% OS, % Sorafenib (n=372) 33-mo rate 40 29% 33% 20 21% 0 0 3 6 9 12 15 18 21 No. at risk NIVO 371 326 273 237 213 189 167 148 SOR 372 330 276 234 198 175 156 133 24 27 30 33 Time, months 36 39 42 45 48 51 130 120 112 102 86 116 98 82 72 59 63 37 42 22 23 10 2 1 0 0 Sangro B, et al. Ann Oncol 2020;31(suppl):abstr LBA-3 161 14.8 (12.1, 17.3) 0.85 (0.72, 1.00); 0.0522 CheckMate 459: long-term efficacy outcomes with nivolumab versus sorafenib as firstline treatment in patients with advanced hepatocellular carcinoma – Sangro B, et al Overall survival by PD-L1 expression Key results (cont.) Tumour-cell PD-L1 expression ≥1% Median OS, mo (95%CI) Nivolumab (n=71) Sorafenib (n=64) 16.1 (8.4, 22.3) 8.6 (5.7, 16.3) HR (95%CI) Tumour-cell PD-L1 expression <1% Median OS, mo (95%CI) 0.80 (0.54, 1.17) Sorafenib (n=300) 15.2 (12.7, 18.1) HR (95%CI) 100 80 80 OS, % 100 60 OS, % Nivolumab (n=295) 16.7 (13.9, 19.4) 40 20 0.84 (0.70, 1.01) 60 40 20 0 0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 Time, months Time, months No. at risk NIVO 71 64 53 43 41 38 32 29 25 24 23 20 16 12 8 SOR 64 53 37 29 28 25 23 22 20 17 15 14 13 12 7 0 0 295 257 216 190 169 148 133 117 104 95 88 81 69 50 34 23 2 300 271 233 199 165 145 128 106 93 78 65 56 45 25 15 10 1 0 0 Sangro B, et al. Ann Oncol 2020;31(suppl):abstr LBA-3 CheckMate 459: long-term efficacy outcomes with nivolumab versus sorafenib as firstline treatment in patients with advanced hepatocellular carcinoma – Sangro B, et al Overall survival by aetiology HCVa Key results (cont.) Nivolumab Median OS, mo (95%CI) HR (95%CI) HBVa (n=87) Sorafenib (n=86) 17.5 (13.9, 21.9) 12.7 (9.9, 16.2) Median OS, mo (95%CI) 0.72 (0.51, 1.02) Uninfected Nivolumab (n=116) Sorafenib (n=117) 16.1 (12.5, 21.3) 10.4 (8.5, 17.3) HR (95%CI) Median OS, mo (95%CI) 0.79 (0.59, 1.07) HR (95%CI) 80 80 80 60 60 60 OS, % 100 OS, % 100 OS, % 100 40 40 40 20 20 20 0 0 0 No. at risk 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 Nivolumab (n=168) Sorafenib (n=168) 16.0 (10.8, 20.2) 17.4 (13.7, 21.3) 0.91 (0.72, 1.16) 0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 Time, months Time, months 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 Time, months NIVO 87 77 67 58 53 48 40 34 29 27 26 25 20 13 8 4 1 0 116 106 86 72 68 56 51 46 42 37 36 33 31 21 14 9 0 168 143 120 107 92 85 76 68 59 56 50 44 35 29 20 10 1 0 SOR 87 74 61 54 43 34 30 25 22 18 17 17 14 7 2 0 0 117 101 77 63 53 50 45 37 33 29 27 24 21 17 8 0 168 154 137 116 101 90 80 70 60 50 37 30 23 13 9 0 5 4 4 1 aPatients could have had active or resolved HBV or HCV infection as a risk factor for HCC Sangro B, et al. Ann Oncol 2020;31(suppl):abstr LBA-3 162 CheckMate 459: long-term efficacy outcomes with nivolumab versus sorafenib as firstline treatment in patients with advanced hepatocellular carcinoma – Sangro B, et al TRAEs Key results (cont.) Fatigue Pruritus Rash AST increased Diarrhoea Appetite decreased Nausea Palmar-plantar erythrodysesthesia syndrome Weight decreased Alopecia Hypertension Dysphonia NIVO grade 3/4 25 15 13 11 11 9 14 9 47 9 26 6 5 11 49 1 1 1 1 1 20 10 11 18 21 12 0 TRAEs, % NIVO grade 1/2 10 20 SOR grade 3/4 30 40 50 SOR grade 1/2 Conclusions ◦ In patients with advanced HCC, 1L nivolumab continued to demonstrate improvements in OS regardless of PD-L1 status or viral aetiology and had a manageable safety profile Sangro B, et al. Ann Oncol 2020;31(suppl):abstr LBA-3 SECOND-LINE SYSTEMIC THERAPY 163 Second-Line Systemic Therapy Regorafenib Nivolumab Cabozantinib Pembrolizumab Nivolumab + ipilimumab Ramucirumab KEYNOTE-240: Pembrolizumab for Patients With Previously Treated HCC ◦ Randomized, double-blind phase III trial of pembrolizumab vs placebo (both with BSC) for pts with advanced HCC with intolerance to or PD on or after sorafenib; Child-Pugh A (N = 413) ◦ Failed to reach prespecified level of statistical significance for OS, PFS in primary analysis (prespecified P = .0174 [OS] and P = .002 [PFS] required) (median f/u 10.6-13.8 mos); updated analysis with additional 18 mos f/u 100 OS (%) 80 60 24-mo rate 28.8% 20.4% 40 20 36-mo rate 17.7% 11.7% Median PFS, Mos (95% CI) Pembrolizumab 3.3 (2.8-4.1) Placebo 2.8 (1.6-3.0) HR (95% CI) 0.70 (0.56-0.89) Nominal P = .0011 100 80 OS (%) Median OS, Mos (95% CI) Pembrolizumab 13.9 (11.6-16.0) Placebo 10.6 (8.3-13.5) HR (95% CI) 0.77 (0.62-0.96) Nominal P = .0112 60 40 24-mo rate 11.8% 4.8% 20 36-mo rate 9.0% 0 0 0 0  0 4 8 12 16 20 24 28 32 36 40 44 48 Mos ORR: pembrolizumab, 18.3%; placebo 4.4% Finn. JCO. 2020;38:193. Merle. ASCO GI 2021. Abstr 268. 164 4 8 12 16 20 24 28 32 36 40 44 Mos CheckMate 040: Nivolumab + Ipilimumab for Advanced HCC ◦ Open-label phase I/II trial of 3 different dosing schemes of nivolumab + ipilimumab for patients with advanced HCC and prior sorafenib treatment; Child-Pugh score A5-A6; ECOG PS 0/1 OS ◦ Dosing: ◦ NIVO1/IPI3 Q3W: nivolumab 1 mg/kg + ipilimumab 3 mg/kg Q3W (4 doses) 100 ◦ NIVO3 Q2W/IPI1 Q6W: nivolumab 3 mg/kg Q2W + ipilimumab 1 mg/kg Q6W NIVO1/IPI3 Q3W (n = 50) NIVO3/IPI1 Q3W (n = 49) NIVO3 Q2W/ IPI1 Q6W (n = 49) 32 (20-47) 31 (18-45) 31 (18-45) ORR, % (95% CI) 80 OS (%) ◦ NIVO3/IPI1 Q3W: nivolumab 3 mg/kg + ipilimumab 1 mg/kg Q3W (4 doses), each followed by nivolumab 240 mg Q2W Median OS, mos (95% CI) NIVO1/IPI3 Q3W 22.2 (9.4-NA) NIVO3/IPI1 Q3W 12.5 (7.6-16.4) NIVO3 Q2W/IPI1 Q6W 12.7 (7.4-30.5) 60 40 20 Median follow-up: 46.5 mos LTFU period 0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 54 Mos Yau. JAMA Oncol. 2020;6:e204564. El-Khoueiry. ASCO GI 2021. Abstr 269. Efficacy and safety of nivolumab + ipilimumab in Asian patients with advanced hepatocellular carcinoma: subanalysis of the CheckMate 040 study Key patient inclusion criteria • Advanced HCC • Sorafenib naïve or progression after or intolerant to sorafenib • Child-Pugh A5 or A6 • HBV, HCV or non-viral HCC • ECOG PS 0–1 (n=71) R 1:1:1 Nivolumab 1 mg + ipilimumab 3 mg q3w x4 (n=50; 34 Asian) Arm B Nivolumab 3 mg + ipilimumab 1 mg q3w x4 (n=49; 27 Asian) Arm C Nivolumab 3 mg q2w + ipilimumab 1 mg q6w (n=49; 29 Asian) PRIMARY ENDPOINTS • Safety, ORR (RECIST v1.1, investigator assessed), DoR Nivolumab 240 mg iv q2w flat dose PD/toxicity Study objective ◦ To evaluate the efficacy and safety of nivolumab + ipilimumab in Asian patients with advanced HCC Arm A SECONDARY ENDPOINTS • DCR, TTR, TTP, PFS, OS Yao T, et al. Ann Oncol 2020;31(suppl):abstr O-5 This talk was presented at the 22nd ESMO WCGC on 1 July 2020 at 18:29 165 Efficacy and safety of nivolumab + ipilimumab in Asian patients with advanced hepatocellular carcinoma: subanalysis of the CheckMate 040 study Asian patients CR SD PD Patients, n (%) PR 60 0 9 (31) 7 (26) 7 6 (21) (18) 2 (6) 24 (49) 20 (40) 40 20 All patients 15 (52) 15 (56) 16 (47) 2 (7) 4 (14) 2 (7) 3 (6) Arm B NIVO3 + IPI1 q3w (n=27) 9 (18) 5 (10) 0 0 Arm A NIVO1 + IPI3 q3w (n=34) 15 (31) 12 (24) 12 (24) 9 (18) 4 (8) 21 (43) Arm C NIVO3 q2w + IPI1 q6w (n=29) Arm A NIVO1 + IPI3 q3w (n=50) Arm B NIVO3 + IPI1 q3w (n=49) Arm C NIVO3 q2w + IPI1 q6w (n=49) ORR, n (%) 8 (24) 9 (33) 9 (31) 16 (32) 15 (31) 15 (31) DCR, n (%) 17 (50) 11 (41) 13 (45) 27 (54) 21 (43) 24 (49) Median DoR, mo (95%CI) 9.8 (7.0, NR) 15.2 (4.2, NR) 11.1 (4.2, NR) 17.5 (8.3, NR) 22.2 (4.4, NR) 16.6 (4.3, NR) Median OS, mo (95%CI) 16.4 (8.6, NR) 11.8 (6.1, 16.4) 11.2 (6.1, NR) 22.8 (9.4, NE) 12.5 (7.6, 16.4) 12.7 (7.4, 33.0) Yao T, et al. Ann Oncol 2020;31(suppl):abstr O-5 Efficacy and safety of nivolumab + ipilimumab in Asian patients with advanced hepatocellular carcinoma: subanalysis of the CheckMate 040 study Key results (cont.) Asian patients Grade 3/4 TRAEs, n (%) Any All patients Arm A NIVO1 + IPI3 q3w (n=33) Arm B NIVO3 + IPI1 q3w (n=27) Arm C NIVO3 q2w + IPI1 q6w (n=29) Arm A NIVO1 + IPI3 q3w (n=49) Arm B NIVO3 + IPI1 q3w (n=49) Arm C NIVO3 q2w + IPI1 q6w (n=48) 17 (52) 7 (26) 8 (28) 26 (53) 14 (29) 15 (31) Pruritus 1 (3) 0 0 2 (4) 0 0 Rash 1 (3) 1 (4) 0 2 (4) 2 (4) 0 Diarrhoea 1 (3) 0 0 2 (4) 1 (2) 1 (2) AST increased 5 (15) 3 (11) 2 (7) 8 (16) 4 (8) 2 (4) 0 0 0 1 (2) 0 0 3 (9) 2 (7) 0 4 (8) 3 (6) 0 Fatigue ALT increased Conclusions ◦ In Asian patients with advanced HCC, nivolumab + ipilimumab demonstrated clinically meaningful responses, particularly in the nivolumab 1 + ipilimumab 3 arm ◦ The safety profile was manageable with no new safety signals observed Yao T, et al. Ann Oncol 2020;31(suppl):abstr O-5 166 167 Checkmate 040 : nivolumab in advanced hepatocellular carcinoma ◦ Nivolumab 3 mg/kg lead to objective response in 16% of the patients using RECIST 1.1 (15% of PR and 1% of CR) ◦ Disease control rate of 68% ◦ Median overall survival of 15 months ◦ Acceptable safety profile ◦ Randomized controlled trial phase 3 comparing sorafenib to ivolumab in advanced HCC (Checkmate 459) El Khoueiry AB, et al. Lancet 2017 168 169 Outcomes for patients with advanced hepatocellular carcinoma (HCC) and Child-Pugh B liver function in the phase 3 CELESTIAL study of cabozantinib vs placebo Study objective ◦ To evaluate the efficacy and safety of cabozantinib in the subgroup of patients with advanced HCC whose liver function had deteriorated to Child-Pugh B by Week 8 Cabozantinib 60 mg/day (n=470) Key patient inclusion criteria • Advanced HCC • Child-Pugh score A • Received prior sorafenib • Progressed after ≥1 prior systemic treatment for HCC • Received ≤2 prior systemic regimens for advanced HCC • ECOG PS 0–1 Stratification • Disease aetiology (HBV, HCV, other) • Geographic region (Asia, other) • Presence of extrahepatic spread and/or macrovascular invasion (EHS/MVI) R 2:1 Placebo (n=237) (n=707) PRIMARY ENDPOINT • OS Loss of clinical benefit / toxicity Loss of clinical benefit / toxicity SECONDARY ENDPOINTS • PFS, ORR, safety El-Khoueiry A, et al. Ann Oncol 2020;31(suppl):abstr SO-9 This talk was presented at the 22nd ESMO WCGC on 1 July 2020 at 19:32 170 Outcomes for patients with advanced hepatocellular carcinoma (HCC) and Child-Pugh B liver function in the phase 3 CELESTIAL study of cabozantinib vs placebo Overall survival Child-Pugh B subgroup Key results Cabozantinib (n=51) Overall Median OS, mo (95%CI) No. of deaths 8.5 (7.7, 12.2) 37 Cabozantinib (n=470) Placebo (n=237) Placebo (n=22) 3.8 (3.3, 4.8) 20 HR 0.32 (95%CI 0.18, 0.58) No. of deaths 10.2 (9.1, 12.0) 317 8.0 (6.8, 9.4) 167 HR 0.76 (95%CI 0.63, 0.92); p=0.005 1.0 Probability of OS 1.0 Probability of OS Median OS, mo (95%CI) 0.8 0.6 0.4 0.2 0 0.8 0.6 0.4 0.2 0 0 6 12 18 24 30 Time, months 36 42 0 6 12 18 24 30 Time, months 36 42 El-Khoueiry A, et al. Ann Oncol 2020;31(suppl):abstr SO-9 Outcomes for patients with advanced hepatocellular carcinoma (HCC) and Child-Pugh B liver function in the phase 3 CELESTIAL study of cabozantinib vs placebo Key results (cont.) Grade 3/4 AEs, % Any Child-Pugh B subgroup (n=51) 71 Overall population (n=467) 68 Fatigue 20 Ascites 14 10 4 AST increased 14 12 Thrombocytopenia 12 3 Palmar-plantar erythrodysesthesia 8 17 Hypertension 8 16 Conclusions ◦ In patients with advanced HCC and Child-Pugh B liver function by Week 8, cabozantinib demonstrated similar outcomes to those of the overall population and had a manageable safety profile El-Khoueiry A, et al. Ann Oncol 2020;31(suppl):abstr SO-9 171 Multiple VEGF-Targeted Therapies Have Activity After Sorafenib: Phase III Data RESORCE CELESTIAL REACH-2 Regorafenib vs placebo Cabozantinib vs placebo (N = 707) Ramucirumab vs placebo 2L, sorafenib-tolerating pts only (N = 573) 2L or 3L (N = 707) 2L, AFP ≥ 400 ng/mL (N = 292) Median OS: 10.6 vs 8.0 mos Median OS: 10.2 vs 8.0 mos Median OS: 8.5 vs 7.3 mos HR: 0.63 (P < .0001) HR: 0.76 OS(P = .005) HR: 0.71 (P = .0199) Regorafenib: multitargeted TKI Ramucirumab: anti-VEGFR2 Ab Cabozantinib: multitargeted TKI Bruix. Lancet. 2017;389:56. Abou-Alfa. NEJM. 2018;379:54. Zhu. Lancet Oncol. 2019;20:282. Evolving Landscape of HCC Nivolumab or lenvatinib Not worse than sorafenib Sorafenib Better than placebo Atezolizumab + bevacizumab Better than sorafenib Anti-VEGF I-O Regorafenib OS vs placebo Pembrolizumab, nivolumab ± ipilimumab Some durable responses Cabozantinib OS vs placebo Ramucirumab OS vs placebo for AFP ≥ 400 ng/mL 172 Systemic treatment sequencing for BCLC Stage C advanced HCC ◦ Targeted first-line therapies ◦ Combination: atezolizumab (PD-L1 inhibitor) + bevacizumab* (VEGF inhibitor) (US only) ◦ Oral multikinase inhibitors: sorafenib and lenvatinib ◦ Targeted second-line therapies ◦ ◦ ◦ ◦ ◦ Multikinase inhibitor: regorafenib Multikinase inhibitor: cabozantinib Anti-VEGFR (AFP ≥400 ng/mL) antibody: ramucirumab PD-1 inhibitors: nivolumab, pembrolizumab Immune therapy Combination: nivolumab + ipilimumab First line atezolizumab + bevacizumab1 Second line AFP ≥400 ng/mL sorafenib lenvatinib regorafenib cabozantinib ramucirumab nivolumab pembrolizumab nivolumab + ipilimumab1 Bruix J, et al. Nat Rev Gastroenterol Hepatol. 2019;16:617-30 Third line cabozantinib 47 173 ADVERSE EFFECTS OF IMMUNOTHERAPY IN HEPATOCELLULAR CARCINOMA - case presentation Dimitar Stefanovski, dr.med. Prof. dr. Janja Ocvirk, dr. med. 2nd International Summer school OCTOBER 2019 • 83 years old female patient; • former smoker, no history of alcohol consumption; • y. 2010 - excision of the submandibular gland (adenoca.), osteoporosis GERD, AMD; • PS WHO 1 • pain under the right costal arch; • abdominal US: tumour formation in the liver; • moderately differentiated, pseudo glandular HCC. 174 • CT thorax + abdomen: hypervascular tumour in the right liver lobe with A – V fistulas, invasion of the middle hepatic vein, without cirrhosis, no signs of distant metastasis. NOVEMBER 2019 • 1st line treatment with Sorafenib • After 1st application: skin toxicity - erythematous skin with macular rash • Continuing the treatment with dose reduced Sorafenib • pain in the palms and soles of the feet, macular rash. DECEMBER 2019 • Sorafenib therapy was terminated prematurely due to side effects. 175 NEXT OPTION? • EMBOLIZATION not possible due to the vascular shunt. • IMbrave 150 tiral results: Patients with unresectable hepatocellular carcinoma have better survival with the combination of atezolizumab and bevacizumab than with sorafenib as first line treatment. 176 JANUARY 2020 • Systemic therapy with Atezolizumab + Bevacizumab. • CT scans showing stable disease. JUNE 2021 • oral mucositis (grade 2) • petechiae, somewhere ulcerated skin with bleeding (grade 1) • productive cough, non purulent sputum (grade 1) • temporarily suspended bevacizumab, continuation with atezolizumab 177 JULY 2021 - HOSPITALIZATION • feeling extremely unwell • cough (grade 2) • bloody diarrhea (grade 2) • ecchymosis, somewhere superficially ulcerated skin with bleeding (grade 1) • negative microbiological analyses • CT scan: PNEUMONITIS 178 TREATMENT • i.v. corticosteroids in high doses 1 mg/kg BW • i.v. Vit K • after 1 week of hospitalization, discharged in stable condition • continuation of corticosteroids therapy p.o. at home THANK YOU FOR YOUR ATTENTION 179 Enrichment of treatment at metastatic NSCLC with PD-L1 overexpression (PD-L1≥50%) Davorin Radosavljevic Institute for Oncology and Radiology of Serbia Belgrade 2nd International Summer School of Oncology Onkološki Inštitut Ljubljana, 08.09.2021. Advent of immunotherapy supstantially changed initial treatment of advanced NSCLC Factors in choosing initial therapy:  Level of PD-L1(≥50%)  Presence or absence of a driven mutation (EGFR, ALK, ROS1, BRAF,etc)  The extent of disease (number and sites of metastases, symptoms)  Squamous vs nonsquamous histology 180 Treatment algorithm for stage IV NSCC, molecular tests negative (ALK/BRAF/EGFR/ROS1), ESMO 2020 Check-point inhibitors, specially targeting PD-1 or PD-L1 have became the cornerstone of 1st line therapy  Monotherapy  Chemotherapy-immunotherapy combinations  Immunotherapy-exclusive regimens Decision-making in this space is complex:  the absence of head-to-head prospective comparisons Paramount for patient selection:  tumor cell PD-L1 expression and histology 181 Immunotherapy today MONOTHERAPY1–6 CheckMate 026 Nivolumab monotherapy CHEMOTHERAPY COMBINATIONS7–13 KEYNOTE-189 IMpower150 Pembrolizumab + cisplatin/ carboplatin + pemetrexed Atezolizumab + bevacizumab + carboplatin + paclitaxel CIT + CIT COMBINATIONS4,14 CheckMate 227 Nivolumab + ipilimumab KEYNOTE-024 Pembrolizumab monotherapy CheckMate 9LA KEYNOTE-407 KEYNOTE-042 Pembrolizumab monotherapy MYSTIC Durvalumab monotherapy Nivolumab + ipilimumab + chemotherapy Pembrolizumab + carboplatin + paclitaxel/nab-paclitaxel IMpower131 CheckMate 227 Atezolizumab + carboplatin + paclitaxel/nab-paclitaxel Nivolumab + chemotherapy MYSTIC Durvalumab + tremelimumab IMpower110 Atezolizumab monotherapy B-FAST* IMpower130 POSEIDON* Atezolizumab + carboplatin + nab-paclitaxel Durvalumab + tremelimumab + chemotherapy Atezolizumab monotherapy IPSOS* Atezolizumab monotherapy IMpower132 EMPOWER-lung 3 Atezolizumab + cisplatin/ carboplatin + pemetrexed Cemiplimab + platinum doublet chemotherapy 1. Carbone, et al. N Engl J Med 2017; 2. Reck, et al. N Engl J Med 2016; 3. Mok, et al. Lancet 2019 4. Rizvi, et al. JAMA Oncol 2020; 5. Spigel, et al. ESMO 2019 (Abs LBA78) 6. Regeneron press release (5 November 2019); 7. Gandhi, et al. N Engl J Med 2018 8. Socinski, et al. N Engl J Med 2018; 9. West, et al. Lancet Oncol 2019 10. Papadimitrakopoulou, et al. WCLC 2018 (Abs OA05.07); 11. Paz-Ares, et al. ESMO IO 2019 (Abs LBA3) 12. Reck, et al. ASCO 2020 (Abs 9501; 13. AstraZeneca press release (28 October 2019) 14. Hellmann, et al. N Engl J Med 2018 EMPOWER-lung 1 Cemiplimab monotherapy *Data not yet made public 5 Tumor PD-L1 expression ≥ 50%: Initial systemic treatment (driven mutation absent or unknown) for patients with squamous or non-squamous NSCLC  Pembrolizumab superior in OS over platinum doublet (KN024)  Atezolizumab superior in OS over platinum doublet (IMpower 110)  Cemiplimab superior in OS over platinum doublet (EMPOWER-Lung1)  An exception, combination doublet chemotherapy with concurrent pembrolizumab : high tumor burden or rapidly progressive disease in which early PD might preclude the option of chemotherapy in 2nd line setting, due to functional decline  Contraindication for immunotherapy (a histology-appropriate platinum doublet): allograft recipiens, myastenia gravis, severe IBD or vasculitis 182 Cochrane Meta-analysis 2020: data from 7 clinical trials comparing the efficacy of PD-L1/PD1 inhibitors to that of chemotherapy in patients with advanced NCSLC (n=5893) Ferrara et al. Cochrane Database Syst.Rev 12. CD13257 (2020)  Anti PD1/PD-L1 antibodies improved OS in patients with tumor cell PDL1≥50% (n=2111) compred with chemotherapy (HR 0.68, 95%CI 0.60-0.76) with a moderate level of certainty  Among never-smokers with PD-L1 expression ≥50%, N=179, no stat.significant difference in OS between PD1/PD-L1 Abs and chemotherapy; current or former smokers had OS advantage with immunotherapy over chemotherapy  No statistically significant difference in OS in PD-L1˂1% or ≥1% Response rate and Survival at Key Timepoints With PD-L1 Blockade vs Chemotherapy in PD-L1 Subgroups: Meta-analysis of Metastatic NSCLC trials J.Man et al. JNCI Cancer Spectrum(2021)5(3):pkabo012  9810 pts in 27 studies, retrospective analysis, systematic research of MEDLINE/EMBASE  In treatment naive patients benefits with PD-L1 blockade over CT were seen in ORR in pts having PD-L1 ≥50%, 2yrOS for PD-L1 ≥1%, 1-yrPFS, 2-yrPFS and 3-yrOS in unselected pts  First-line PD-L1 blockade compared with CT: Higher ORR, 2-yrPFS and 3-yrOS if PD-L1 was 50% or greater Lower ORR, higher 2-yrPFS and similar 3-yrOS if PD-L1 was 1-49% Lower ORR, similar 1-yrPFS and lower 2-yrOS if PD-L1 was less than 1%  In previously treated patients, PD-L1 blockade demonstrated similar or superior outcomes in all PD-L1 subgroups 183 Outcomes to first-line pembrolizumab in patients with non-small-cell lung cancer and very high PD-L1 expression Aguilar EJ. Et al. Ann Oncol 2019. Multicentric retrospective analysis 187 patients, ORR 44.4%, mPFS 6.5mo, mOS NR  PD-L1 50-89% N=107pts, ORR 32.7%, mPFS 4.1mo, OS 15.9mo  PD-L1 90-100% N=80pts, ORR 60%, mPFS 14.5mo, OS NR Implication: treatment selection, clinical trial intepretation and design Does immuno-chemotherapy and imuno-immunotherapy provide additional benefit beyond that of pembrolizumab or atezolizumab monotherapy in PD-L1≥50% population? 24 months OS: 50% pembrolizumab mono in KEYNOTE 024 trial 52% pembrolizumab+chemotherapy in PD-L1≥50% subgroup of KEYNOTE189 48% with nivolumab+ipilimumab in CheckMate227 trial. Immuno-chemo and immuno-immuno combination are associated with predictable but higher risk of toxicities, while mono anti-PD1/PD-L1 provide impressive levels of tolerability compared to chemotherapy AntiPD1/PD-L1 antibodies as monotherapy for most patients 184 Whom we should offer combo immuno-chemotherapy in PD-L1≥50% population? Imminent need for cytoreduction:  substantial tumor burden,  rapidly progressive disease and/or  severe disease related symptoms Numerically higher ORR than PD-1/PD-L1 monotherapy Rapid cytoreduction produced by combo might also attenuate the early decrements in OS seen in patients receiving monotherapies Only 51% of patients with PD on pembrolizumab in KN024 and 38% in experimental arm of KN042 received subsequent therapy Mono vs combo approach ORR in patients with PD-L1 expression > 50% Combo therapy 80% 80% 70% 70% Objective Response Rate (%) Objective Response Rate (%) Monotherapy 60% 46% 50% 39% 40% 40% 30% 20% 10% 69% 62% 60% 60% 50% 50% 40% 30% 20% 10% 0% 0% KN 024 KN 042 IMP 150 IMP 110 KN 189 KN 407 Brahmer et al ESMO 2020 KEYNOTE-024 5 year OS update, Gray et al WCLC 2020 KEYNOTE-189 4 year OS update, Cho et al WCLC 2020 KEYNOTE-042 3 year OS update, Herbst, et al. WCLC 2020 Abs FP13.03, Reck et al ASCO 2021 (Abs 9000), Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non–small-cell lung cancer. N Engl J Med 2018;379:2040-51. DOI: 10.1056/NEJMoa1810865 185 CM 9LA Addition of chemo to immunotherapy does not add benefit in nonsquamous mNSCLC with high expression of PD-L1 (except in patients with no smoking history) S.Peters et al. ESMO Virtual Pnenary, April 8, 2020. Observational study, Flatitron Health database, 520 pts, PS 0-2, PD-L1≥50%, pembrolizumab mono (N=351) or plus paclitaxel-carboplatin (N=169) Mono arm: higher proportion of poor prognostic baseline characteristics (age, extent of disease) Median OS 22.1mo vs 21.0mo p=0.63 HR 1.03, PFS 11.5 vs 10.8mo No smoking population: combo reduced risk by 75%, P=0.02 and 60%,P=0.04 Liver and brain metastases: outcomes were similar Invited discutant F.Cappuzzo, ESMO 2020. About 30% of patients in phase III clinical trials have better survival outcome with chemotherapy than with single-agent CPI. In most relevant trials mOS with single-agent was less than two years, but more than two years in combo approach, ORR were also higher Cost: toxicity – grade 3-5 in appr. 60% with combo, compared to 20% treated with single agents Combo certainly in never-smokers Not all patients with PD-L1 are equall (Pts with≥90% are extremly sensitive to immunotherapy) 186 Mono imunotherapy options in 1L NSCLC ORR,DoR, PFS, OS in PD-L1 >50% population 39% 40% 30% 20% 25 20 15 10 10% 5 0% KN 042 IMP 110 7.7 8 6.5 7 26.3 6 5 4 3 2 25 20 20.2 KN 042 IMP 110 20 15 10 5 1 0 0 KN 024 30 8.1 Overall survival (months) 27.3 30 46% 40% 29.1 9 Progression free survival (months) 35 60% DoR (months) Objective Response Rate (%) 38.9 40 70% 50% 10 45 80% KN 024 KN 042 0 KN 024 IMP 110 KN 042 Brahmer et al ESMO 2020 KEYNOTE-024 5 year OS update, Cho et al WCLC 2020 KEYNOTE-042 3 year OS update, Herbst, et al. WCLC 2020 Abs FP13.03, Herbst, et al. N England J Med 2020 Combo and hyperprogression A subset of patients have a paradoxical acceleration in tumor growth and clinical deterioration upon initiation of therapy The incidence of hyperprogression in previously treated NSCLC ranges from 8 to 21% In treatment-naive NSCLC receiving anti-PD1/PDL1 as monotherapy the incidence was 16% This effect can largely be avoided using first-line chemo-immunotherapy combinations 187 IMP 110 KN 024 Therapy duration Up to 2 years in KEYNOTE and CheckMate 227 trials Indefinite maintenance in IMpower119, IMpower 130 and IMpower150 Results of KN158 randomized study favored longer duration than a 1 year fixed dose (small sample, lack of alternative fixed dose, such as 2 years) In KEYNOTE 024 39 of 154 pts (25%) completed 2 years pembrolizumab, and 3-year landmark OS from completion was 81% of These data suggest that most patients completing 2 years of treatment continue to derive long-term benefit, despite therapy cessation The decision to discontinue maintenance in the absence of substantial toxicities or disease progression should be individualized: whether therapy beyond 2 years improves outcome remains largely unclear Post-progression outcome of NSCLC patients with PD-L1 expression ≥50% receiving first-line single-agent pembrolizumab in a large multicentre real-world study Cortellini A. et al, Eur J Cancer 2021. 974 patients were included, median follow-up 22.7months Median OS 15.8 months (95% CI:13.5-17.5; 548 events) 55.9% had not received any further treatment, 52.9% died Patients who did not receive post-PD therapies were: older (p=0.0011), with a worse PS (p˂0.0001) and were on corticosteroids prior to pembrolizumab (p=0.0024); At disease progression, 29.2% received a switched approach, 14.9% received pembrolizumab beyond PD, either alone (9.4%) or in combo with local ablative treatments (5.5%): OS 13.9 vs 7.8mo (p=0.0179) 35.5% received second-line systemic treatment (sign.higher proportion of age under 70, poorer PS, having CNS and liver mets) 188 Real-world study (Cortellini 2021) and KN024: Pts with PDL1≥50% Median OS 15.8mo vs 26.3mo Not unsuprising: higher proportion of patients with adverse prognostic factors (PS ≥2, receiving corticosteroids, older than 70years) Patients in clinical studies are highly selected for lower co-morbidity burden Inferior outcome: patients with poor baseline PS, particulary if related to disease burden; 55.9% did not receive any further treatment; Real-world study (Cortellini 2021) and KN024: Pts with PDL1≥50% Older pts, PS ≥ 2, receiving systemic corticosteroids are at higher risk of lifethretening progressive disease – thus, treatment sequencing approach ICI-CT doublet is unlikely to be completed Oligoprogression and pembrolizumab beyond PD: the best post-PD outcome: pembrolizuimab beyond PD + local ablative treatment Among patients who reach a second-line treatment, PS still remains th major determinent of clinical outcome 189 Take home messages – mNSCLC with PD-L1≥50% Decision-making in this space is complex: the absence of head-to-head prospective comparisons Paramount for patient selection: tumor cell PD-L1 expression and histology AntiPD1/PD-L1 patients antibodies as monotherapy for most Not all patients with PD-L1 are equall: patients with ≥ 90% are extremly sensitive to immunotherapy, non-smokers are for combo approach or chemotherapy Take home messages – mNSCLC with PD-L1≥50% The decision to discontinue maintenance in the absence of substantial toxicities or disease progression should be individualized: whether therapy beyond 2 years improves outcome remains largely unclear Hyperprogression can largely be avoided using first-line chemo-immunotherapy combinations Toxicity – grade 3-5 in appr. 60% with combo, compared to 20% treated with single agents 190 Clinical choices in metastatic NSCLC without actionable oncogenic driver mutations regardless of PD-L1 status Marko Jakopović, MD, PhD Associate Professor Zagreb Medical School Department of Respiratory Medicine University Hospital Centre Zagreb Disclosures Speakers fees: AstraZeneca, Roche, Boehringer Ingelheim, Eli Lilly, Berlin Chemie, Sandoz, Novartis, MSD, Novartis Oncology, Abbott Advisory boards: AstraZeneca, Roche, BoehringerIngelheim, Eli Lilly, MSD, Novartis Oncology 191 Overview How to choose treatment options in metastatic NSCLC in patients without driver mutation?  Immunotherapy vs chemotherapy? Nivolumab vs docetaxel in previously treated squamous NSCLC Brahmer J et al. N Engl J Med 2015;373:123-135. 192 Pembrolizumab in treatment-naive highly positive PD-L1 NSCLC patients Reck M et al. N Engl J Med 2016; 375:1823-1833. Atezolizumab was superior to docetaxel regardless of PD-L1 expression in NSCLC Rittmeyer et al. Lancet 2016. 193 Overview How to choose treatment options in metastatic NSCLC in patients without driver mutations?  Immunotherapy vs chemotherapy? The winner is immunotherapy!!!!!  Line of treatment of immunotherapy?  Monotherapy vs combination therapy (CT+IO, IO+IO)?  Does histology matter?  Open questions  Conclusions Line of treatment of immunotherapy? 194 Nivolumab in heavily preatreated patients Brahmer JR et al. N Engl J Med 2012;366:2455-2465. Second-line immunotherapy in NSCLC 195 KeyNote-024: First-line pembrolizumab in PD-L1 highly positive patients Overview How to choose treatment option in metastatic NSCLC in patients without driver mutations?  Immunotherapy vs chemotherapy? The winner is immunotherapy!!!!!  Line of treatment of immunotherapy? The winner is first – line immunotherapy!!!!  Monotherapy vs combination therapy (CT+IO, IO+IO)?  Does histology matter?  Open questions  Conclusions 196 Monotherapy vs combination therapy (CT+IO, IO+IO)? Keynote-042: Pembrolizumab vs Chemotherapy in PD-L1 positive NSCLC 197 Keynote-042: Overall survival in ITT population Mok TS et al. Lancet 2019 Overview How to choose treatment option in metastatic NSCLC in patients without driver mutations?  Immunotherapy vs chemotherapy? The winner is immunotherapy!!!!!  Line of treatment of immunotherapy? The winner is first – line immunotherapy!!!!  Monotherapy vs combination therapy (CT+IO, IO+IO)? In patients with PD-L1 expression <50% the winner is combination therapy!!!!  Does histology matter?  Open questions  Conclusions 198 Does histology matter? 18 199 IMower131: Study Design Maintenance therapy (no crossover permitted) Stratification factors: • Sex • PD-L1 IHC expression • Liver metastases N = 1021 R 1:1:1 Arm B Atezolizumab + Carboplatin + Nab-Paclitaxel Until PD per RECIST v1.1 or loss of clinical benefit Atezolizumab 4 or 6 cycles Arm C (control) Carboplatin + Nab-Paclitaxel 4 or 6 cycles Co-primary endpoints Best Supportive Care Until PD per RECIST v1.1 Secondary endpoints • Investigator-assessed PFS per RECIST v1.1 (ITT) • OS (ITT) a Atezolizumab 4 or 6 cycles Survival follow-up Arm A Atezolizumab + Carboplatin + Paclitaxel Stage IV squamous NSCLC • Chemotherapy naivea • ECOG PS 0 or 1 • Any PD-L1 IHC status • PFS and OS in PD-L1 subgroups • ORR, DOR; safety Atezolizumab 1200 mg IV q3w; carboplatin AUC 6 IV q3w; nab-paclitaxel 100 mg/m2 IV qw; paclitaxel 200 mg/m2 IV q3w. Patients with a sensitising EGFR mutation or ALK translocation must have disease progression or intolerance to treatment with ≥ 1 approved targeted therapies. Testing for EGFR mutation or ALK translocation was not mandatory. b PD-L1 expression was evaluated using the VENTANA SP142 IHC assay. Jotte R, et al. IMpower131 PFS Analysis. 200 https://bit.ly/2snPEzb Progression-Free Survival (%) INV-Assessed PFS in the ITT Population (Arm B vs Arm C) Median PFS (95% CI), mo Arm B: Atezo + CnP Arm C: CnP 6.3 (5.7, 7.1) 5.6 (5.5, 5.7) HRa (95% CI) P value 0.71 (0.60, 0.85) 0.0001 12-month PFS Minimum follow-up, 9.8 mo Median follow-up, 17.1 mo 24.7% 12.0% Time (months) No. at risk Data cutoff: January 22, 2018. INV, investigator. a Stratified HR. https://bit.ly/2snPEzb Jotte R, et al. IMpower131 PFS Analysis. CheckMate 9LA: NIVO + IPI + 2 cycles of chemo in 1L NSCLC Overall survival by histology NSQ NSCLCa SQ NSCLCb NIVO + IPI + chemo (n = 246) 17.0 11.9 (14.0–NR) (9.9–14.1) 0.69 (0.55–0.87) Median OS, mo (95% CI) HR (95% CI) 100 83% Chemo (n = 246) 100 80 76% 80 63% 60 NIVO + IPI + chemo 50% 40 OS (%) 73% OS (%) NIVO + IPI + chemo Chemo (n = 115) (n = 112) 14.5 9.1 Median OS, mo (95% CI) (13.1–19.4) (7.2–11.6) HR (95% CI) 0.62 (0.45–0.86) 64% 71% 60 NIVO + IPI + chemo 40 40% Chemo 20 Chemo 20 0 0 0 3 6 9 12 204 180 170 152 154 122 No. at risk 224 223 15 18 Months 107 87 62 53 21 24 27 20 18 6 9 0 0 30 0 0 201 0 115 112 3 6 9 12 102 96 88 80 80 56 73 44 15 18 Months 46 29 24 14 21 24 13 8 4 2 27 1 0 30 0 0 Overview How to choose treatment options in metastatic NSCLC in patients without driver mutations?  Immunotherapy vs chemotherapy? The winner is immunotherapy!!!!!  Line of treatment of immunotherapy? The winner is first – line immunotherapy!!!!  Monotherapy vs combination therapy (CT+IO, IO+IO)? In patients with PD-L1 expression <50% the winner is combination therapy!!!!  Does histology matter? Combination treatment is superior to chemotherapy alone regardless of histology subtype!!!!  Open questions  Conclusions Sorry…. I still have to talk about patients wit metastatic NSCLC and driver mutations 202 Check-point inhibitors in EGFR mutated lung cancer Lee CK et al. J Thorac Oncol 2016; in press In EGFR/ALK+ NSCLC patients, OS benefit seen with combination of bevacizumab + atezolizumab + chemotherapy EGFR/ALK+ EGFR/ALK+ Bev + atezo + CP* vs Bev + CP Atezo + CP vs Bev + CP 100 Bev + atezo + CP Bev + CP HR†=0.54 (95% CI: 0.29–1.03) 80 Overall survival (%) Overall survival (%) 100 60 40 20 0 17.5 mo NE HR†=0.82 (95% CI: 0.49–1.37) 80 60 40 20 0 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 Time (months) Atezo + CP Bev + CP 17.5 mo 21.2 mo 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 Time (months) OS benefit in EGFR/ALK+ patients was observed despite lower PD-L1 expression in these patients *One patient had EGFR exon 19 deletion and also tested ALK positive per central lab; †Unstratified HR; Data cut-off: 22 January 2018 Socinski, et al. ASCO 2018 (Abstract 9002); Kowanetz, et al. AACR 2018 (Abstract CT076) 203 26 Mono-immunotherapy vs combination therapy? Mono vs combination of IO plus chemotherapy – what to choose? 204 Overview How to choose treatment options in metastatic NSCLC in patients without driver mutations?  Immunotherapy vs chemotherapy? The winner is immunotherapy!!!!!  Line of treatment of immunotherapy? The winner is first – line immunotherapy!!!!  Monotherapy vs combination therapy (CT+IO, IO+IO)? In patients with PD-L1 expression <50% the winner is combination therapy!!!!  Does histology matter? Combination treatment is superior to chemotherapy alone regardless of histology subtype!!!!  Open questions? In patients with driver mutations combination of IO+bevacizumab+CT is an option!!!! In patients with PD-L1<50% combination therapy is better option!!!! Some questions open in PD-L1≥50% about monoIO vs combination!!!!  Conclusions Instead of conclusions 205 Long-term survival with mono-immunotherapy Long-term survival with combination therapy 206 Systemic treatment of head and neck cancer – what's new in the old Assist. Professor Cvetka Grašič Kuhar, MD, PhD Institute of Oncology Ljubljana, Slovenia Main question: comparing curative locoregional treatment (LRT) with LRT+Cht Which treatment is the most effective? 207 Conclusions regarding systemic treatment in SCHNC • Treatment of stage III/IV disease • Cisplatin based CRT remained the standard of care in cisplatin-fit patients • There is evidence from 2 phase III trials that weekly cisplatin could be delivered instead of high dose cisplatin, efficacy is similar, treatment is less toxic • Addition of cetuximab or ICI does not improve outcome • Replacing cisplatin with cetuximab or ICI is less effective (less locoregional control) • Xevinepant may further increase treatment efficacy • Recurrent/metastatic disease • Cisplatin sensitive: • ICI (Immune checkpoint inhibitor pembrolizumab) in I. line of SCHNC • with Chemotherapy (CPS≥1) or as monotherapy (CPS ≥ 1/, >20) • Extreme regimen: CPS<1 or contraindication for ICI • Cisplatin resistant : ICI monotherapy, mono-chemotherapy 208 Updated results of patients with recurrent or metastatic head and neck cancer treated with nivolumab between January 2018 and March 2020 in Slovenia In total, 27 patients with head and neck cancers were treated with nivolumab in this period. The cohort consisted almost entirely of male patients (96%) and the median age was 59 years. Most of these patients were smokers (89%), and their performance status was 0-1 in 88.9%. The majority had pharyngeal cancer (63%) of which 3 were HPV positive. Almost a third of patients were heavily pre-treated, having received three or more lines of systemic therapy. Cetuximab was a part of this treatment in 44% of patients. There were in total 14 immune-related adverse events occurring in 8 patients in total, of which there was only one case of grade 2-3 event (bullous pemphigoid), while other 13 events were all grade 1. Only one patient received concurrent radiotherapy (60Gy to parastomal recurrence). At the end of the updated observation period there were 22 deaths in total. Median overall survival from the first nivolumab application was 10.7 months (95% CI 1.7-19.7) which is comparable to the CheckMate 141 results.1 One-year overall survival rate was 48.1% (95%CI 29.3-66.9). Univariate analyses of possible predictive variables with log-rank test, namely the impact of presence of distant metastases, immune-related adverse events, neutrophil to lymphocyte ratio, and impact of concurrent antibiotics treatment on overall survival, found no statistically significant correlation. Individual patients’ timelines from diagnosis to death or to last follow up are presented in figure 1. Overall, we can conclude that treatment with nivolumab has been shown to be safe also in Slovenian patients with recurrent or metastatic head and neck cancer. Due to the small number of patients, it is difficult to align the outcomes with the results of previous studies, but in general results are comparable. Figure 1. Individual patients’ timelines from diagnosis to death or to last follow up References 1. Ferris RL, Blumenschein G, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med 2016;375(19):1856– 67. 209 Outcome of patients with recurrent/metastatic squamous cell head and neck cancer treated with platinum-based chemotherapy with or without cetuximab in real-world practice Tina Zupančič, prof.dr. Branko Zakotnik, doc.dr. Cvetka Grašič Kuhar • A retrospective analysis of patients with R/M SCHNC treated at the Institute of Oncology Ljubljana between April 2008 and May 2018. • A total of 67 patients: 34 patients received the PF (platinum, 5-FU), 33 the PFE (PF + cetuximab). • Inclusion criteria for the PF and PFE protocols: First-line therapy for R/M SCHNC, PS 0-2, adequate haematologic, renal and liver function, approved reimbursement for cetuximab (for PFE only). • Exclusion criteria for the PE and PFE protocols: Patients with nasopharyngeal carcinoma, PS >2. • Exclusion criteria for PFE only: Infusion reaction to cetuximab grade >2, prior treatment with cetuximab, known allergy to bee or wasp venom grade >2,bulky tumour in the oropharynx or larynx. • The primary aim: to compare the PSF and OS in the routine clinical setting with outcomes in a randomized trial and to identify possible prognostic factors for PFS and OS. • The secondary aim: to assess the tolerability. Conclusions • PFE regimen has improved OS (but not PFS) when compared to the PF regimen (median PFS 7.1 vs 6.6; median OS 11.5 vs 9.6 months). • Objective response to therapy, good nutritional status and possible further treatment after progression have better prognosis (prognostic factors for OS). • Our results regarding OS are comparable to the EXTREME study. • Similar median PFS and OS with PFE were reported by other real-world studies. • OS of our patients is better than real-world global OS. • No differences in diarrhoea, hypomagnesaemia, infections and febrile neutropenia. • High mortality (13.4%). • PFE still represents the optimal 1st line therapy for fit patients with PD-L1 negative R/M SCHNC (20-30%) or with contraindication for anti-PD-L1 inhibitors and as 2nd line therapy. 210 Management of cancer of unknown primary in the molecular era 2nd Summer School in medical oncology, September 8th Erika MATOS,MD, PhD Kaja CANKAR, MD Definition • CUP is biopsy-proven malignancy for which the anatomic origin at the time of presentation remains unidentified in spite of a detailed history, physical examination and a thorough diagnostic work-up. • CUP is a heterogeneous group of metastatic tumors, which share some common features: • • • • • the ability of an early dissemination, clinical absence of the primary site, aggressive behaviour, unpredictable metastatic pattern, poor response to conventional systemic cytotoxic therapy. Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. 211 Basic diagnostic-work-up in CUP (ESMO guidelines) • Patient’s history • history of previous biopsies, spontaneously regressing lesions and family history • Physical examination • Including rectal and breast examination. • Good quality tissue sample (ESENTIAL!): • meticulous immunohistochemistry. • Basic blood and biochemical analyses. • CT of the chest, abdomen and pelvis. • Mammography in women. Diagnostic strategy should take in account the natural behaviour of the disease and the expected duration of survival based on extent of the disease and PS. Difficult and time-consuming diagnostic studies should not compromise patients' quality of life. Ann Oncol 2015; 26(Suppl 5): v133-138. Additional diagnostic-work-up in CUP (1) • Additional procedures should be sign-, symptom-, lab. abnormalities guided. • Breast MRI: in patients with isolated axillary lymph node metastases and suspected occult primary breast carcinoma after negative mammography and sonography results. • Broader use of MRI in CUP diagnostics is questionable. • Endoscopy: if the patient has symptoms or relevant signs. • FDG-PET imaging in CUP diagnostics: • in patients with cervical lymphadenopathy of primarily squamous histological subtype. • PET-CT is useful (not been prospectively studied): • Patients presenting with solitary metastatic disease who are candidates for curative loco-regional treatment in purpose to exclude occult metastases before extensive surgery, • Patients with known severe iodine dye allergy • Patients with predominant bone disease who would otherwise require either multiple MRIs or bone scans to evaluate response to therapy. Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. 212 Additional diagnostic-work-up in CUP (2) • Serum tumour markers have no proven prognostic, predictive or diagnostic assistance. • Increased values of some tumour markers may help in guiding further diagnostics: • Beta human chorionic gonadotropin (beta-HCG) and alpha-fetoprotein (AFP): • in patients with midline tumour masses with undifferentiated histology. • Prostate Specific Antigen (PSA): • in men with adenocarcinoma and predominantly bone disease. Unfortunately, most tumour markers (CEA, CA125, CA19-9 and CA15-3) are not specific and thus are not helpful in searching for the site of primary tumour. Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. Clinical presentation of patients with CUP? • There is no unique clinical picture. • The majority of patients presents with symptoms and signs of metastatic disease. • There are patients with only or manly liver metastases, with lymph node metastases in mediastinal or retroperitoneal region, with axillary lymph nodes, with cervical lymph nodes, with peritoneal disease, with malignant ascites, with lung disease only or pleural effusion only, bone only disease or metastases to CNS only, although more often as a part of disseminated disease. • Clinical presentation depends on number of metastatic lesions and their distribution. • The majority of patients has metastatic disease in more than one organ, the most often in liver, lung, bone and lymph nodes. Ann Oncol 2015; 26(Suppl 5): v133-138. 213 How can pathologist help? (1) • Challenging work! Direct communication between clinician and pathologist is crucial. • The trend across all cancer types is personalized medicine (CUP seem ideal candidate). • Core biopsy is preferred over fine needle aspirate specimen. • Light microscopy (LM): the tissue specimen (paraffin sections stained with eosin and haematoxylin) • Based on established cytological criteria, the pathologist usually can classify the tumors into broad groups: • Carcinoma (5% SSC)OR adenocarcinoma (60%), • Sarcoma, • Lymphoma. • Some specimens will lack any cytological distinguishing features: • undifferentiated malignancy (35%). Ann Oncol 2015; 26(Suppl 5): v133-138. How can pathologist help? (2) • IHC: significant role in the workup of CUP • Define tumour lineage by using peroxidaselabelled antibodies against specific tumour antigens. • Have to be directed in terms of clinical and radiological patient's data. • Random use of large numbers of tissue markers is rarely helpful. • Staining for different CK (components of cytoskeleton of epithelial tissue) may be very helpful. • Commonly used staining for CK7, 20, 5 and 6. • From the pattern of theirs' expression, the most likely site of origin can be identified. The method has limitations: • the majority of tissue markers are not specific for one organ • no pattern is 100% specific, • the absence of markers does not exclude the origin in certain organ/tissue. Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. 214 How can pathologist help? (3) • • • • The value of some antibodies, such as AFP, beta-HCG, PSA is well established. Some stainings are organ/tissue specific, such as ER, leukocyte common antigen. The absence of certain marker does not exclude the origin in that organ/tissue. The majority of tissue markers are not specific for one organ. Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. How can pathologist help? (4) • Novel molecular approaches in CUP evaluation: • Gene expression profiling tests (GEP) to identify the tissue of origin (ToO) • Comprehensive genomic profiling tests (CGP) to find treatable (clinically relevant) genomic aberrations (GA) Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. 215 Gene expression profiling • Methodology: RT-PCR or microarrays evaluating the expression od different genes • Several assays on the market (evaluating from 10 to 92 and more genes) • Tested in a randomized phase III trial (GEFCAPI 04) Gene expression tests • Are molecular cancer classifiers that help identify the site of origin for cancers with indeterminate, uncertain, or differential diagnoses. • CancerTYPE ID® uses real-time RT-PCR to measure the expression of 92 genes in the patient’s tumor and classifies the tumor by matching the gene expression pattern of the patient’s tumor to a database of known tumor types and subtypes, encompassing 50 tumor types (not FDA approved). • Tissue of Origin Test® is based on Affymetrix microarray. It compares the expression for 2000 genes in a patient‘s tumor with a panel of 15 know tumor types that were diagnosed according to current clinical and pathological practice (FDA approved in June 2010). 216 GEFCAPI 04 (Study description) • Phase III trial of empiric chemotherapy with cisplatin and gemcitabine (GC) or systemic treatment tailored by molecular gene expression analysis in patients with carcinomas of an unknown primary (CUP) site • TOO (Pathwork; n=21) or CancerTYPE ID (Biotheranostics; n=222); N=243 • Primary endpoint: PFS • Secondary endpoints: • OS • PFS in patients with cancers likely insensitive to GC Stratification: PS, LDH level Ann Oncol30; 2019 (Suppl 5): v851–v934. GEFCAPI 04 (Results) • Primary cancers most often reported: pancreatico-biliary (19%), SCC (11%), kidney cancer (8%), lung cancer (8%) • 91/123 (arm B): tailored treatment • PFS: 5.3 mos (arm A) vs 4.6 mos (arm B); HR 0.95 (0.72-1.25); p=0.7 • OS: 10.0 mos (arm A) vs 10.7 mos (mos) (arm B); HR 0.92 (0.69-1.23) • 60 pts with suspected cancers likely insensitive to GC Conclusion: In GEFCAPI 04, using a molecular test followed by tailored systemic treatment did not improve outcomes of pts with CUP. Ann Oncol30 (Suppl 5); 2019: v851–v934. 217 Comprehensive genomic profiling • Methodology: NGS • Ongoing CUPISCO trial: A Phase II Randomized Study Comparing the Efficacy and Safety of Targeted Therapy or Cancer Immunotherapy Versus Platinum-Based Chemotherapy in Patients With Cancer of Unknown Primary Site (NCT03498521) https://clinicaltrials.gov/ct2/show/NCT03498521. Ross JS et al. The Oncologist 2021;26:e394–e402. CUPISCO (Study description) • Aim: to determine the efficacy and safety of targeted therapies and cancer immunotherapies for patients with a subset of CUP syndrome. • The selection of therapies is based on results from CGP (FMT*) • N=790; F II, global, 162 sites participate • After 3 cycles of induction platinumbased ChT doublets the responders are randomized to experimental arm (targeted therapy tailored to the molecular profile) or continue with standard ChT. The non-responders go directly to molecular guided therapy. • • • https://cup-syndrome.com/en/home/cupisco-study.html *FMT: Foundation Medicine tissue or liquid Test 218 The primary endpoint: PFS or death from any cause. The secondary endpoints: OS, RR, CBR, safety…. Estimated date of competition: 2023. Do we (currently) have effective drugs for CUP patients? a responsive subset: an unresponsive subset: favourable prognostic subset poor prognostic subset • About 20% of CUP patients • Have histopathology, biomarkers and clinical presentation consistent with specific tissue of origin • Should be treated with primary-specific therapy corresponding to most likely primary site • About 80% of CUP patients • No type of ChT prolonged survival in these patients Int J Cancer 2014; 135, 2475–81. Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. Favourable prognostic subset • Traditionally defined favourable subset: • • • • • women with isolated axillary adenopathy, women with serous papillary peritoneal carcinomatosis, squamous cell carcinoma involving mid-high cervical lymph nodes, poorly as well as well-differentiated neuroendocrine carcinoma, poorly differentiated and undifferentiated carcinoma (extra gonadal germ cell cancers), • men with blastic bone metastases and elevated PSA • patients with single, small and potentially resectable tumours • Newly identified favourable CUP subset: • patients who look like CRC (CK 20 pos, CK 7 neg, CDX pos), should be treated as patients with advanced CRC (expected RR around 50% and mOS up to 3 years) Abeloff’s Clinical Oncology (6th Edition) 2020; Cancer of Undefined Site of Origin 1694-702. 219 Unfavourable prognostic subset (1) • Sensitivity to chemotherapy is modest. • Include the patient in clinical trial (if an option). • Do the CGP to identify potentially treatable, clinically relevant GA and treat accordingly. • In many countries expensive molecular assays are not available or not covered by insurance. • Targeted drugs and check point inhibitors are not covered by insurance. • At the time being we have no prove that such approach really influence patients' survival. Data from well designed clinical trials are necessary (are ongoing). Unfavourable prognostic subset (2) • The majority of patients from this subset have poor prognosis. • At presentation, two-thirds of patients have metastatic lesions in two or more visceral sites (most often liver, lung, lymph nodes and/or peritoneum). • Patients are often in poor performance status. • • • • For many of these patients BSC is the best option. For selected patients empiric chemotherapy is justified. Cisplatin or taxane-based doublets have been used, with little impact on survival. Patients and relatives have to be informed that expected RR to ChT is only 20% to 30% and expected mOS not more than 9 to 11 mos. This might influence theirs' decision about treatment. NCCN guidelines 220 Burning questions • 1. Does site-specific therapy determined by GEP improves outcome in patients with CUP? • According to the result of GEFCAPI 04 study: NO • The study has limitations! • 2. If genomic profiling identifies a targetable mutation in a tissue sample of CUP and the patients receives targeted therapy, is the outcome improved compared to standard chemotherapy? • It seems logical, but it is not necessary. • A good lesion in terms of this is BRAF mutation. It can be effectively treated if CUP has metastasized from melanoma but not from colon cancer. • The study is underway. Summary and assessment algorithm (1) • Does molecular profiling assay increases accuracy of identifying the primary site? • Most probably yes. • Does molecular profiling (CGP) help us in directing effective targeted treatment? • We are not certain yet. Studies are ongoing. • Does identification of primary site based on molecular assays (GEP tests) and accordingly directed therapy improves patient survival? • According to the results we have: NO. • How to proceed knowing all these in current clinical practice? 221 Summary and assessment algorithm (2) • Try to search for primary site: clinically, by IHC, imaging, endoscopy studies. • Rule out potentially treatable or curable tumours (breast cancer, germ cell tumour, lymphoma) • Select, which clinic-pathological entity the patient belongs to: • If to favourable prognostic subset: • treat accordingly • If to poor prognostic subset: • • • • Have a profound discussion with the patient and relatives about the disease Evaluate patient performance, bear in mind patient’s condition and preferences Choose either empirical ChT or BSC Do CGP, look at targetable, clinically relevant GA and treat the patient on the bases of the results of this test. Of note: without big evidence so far that this approach is beneficial. • The best option: include the patient in a clinical study if available. Conclusions • CUP is a heterogeneous disease with poor prognosis. • It is mandatory to establish to which prognostic group the patient belongs to. • In patients belonging to a favourable prognostic subset long-term survival can be achieved with appropriate treatment. • Patients classified to unfavourable prognostic subset have to be informed about benefits and disadvantages of empiric therapy. For many patients with widespread disease on first presentation and poor PS BSC is the best option. • Novel approaches (searching for clinically relevant GA) are promising, present a fundamental shift in the paradigm of treatment of cancer patients from tissue-specific to individual, patient/tumour customized treatment, directed according to tumour specific GAs. 222 2nd Summer School in Medical Oncology Precision oncology- Are we there yet? 7. - 10. September 2021 Systemic treatment of Ewing sarcoma Where do we stand? Mojca Unk, MD,MS Institute of Oncology Ljubljana Department of medical oncology James Stephen Ewing 1866- 1943 American pathologist the first Professor of pathology at Cornell University 1921: discovery of a form of bone cancer later named Ewing's sarcoma 223 Introduction • a small, blue, round cell sarcoma (RCS) gene fusion involving a member of the FET family and a member of the ETS family of transcription factors (definitive diagnosis) • the 3rd most common bone sarcoma (incidence: ~0.1/100,000/year) • children and adolescents (rarely adults) • median age at diagnosis: 15 y • male predominance • extremity bones (50%), pelvis, ribs and vertebra (any bone, soft tissue) Rizk et al. Pharmgenomics Pers Med. 2019 Outcome • historical series: 5-year survival <10 % (micrometastatic disease) • current recommended multimodal approaches: • Localised disease 5-year survival is ∼60 % - 75 % • Metastatic disease 5-y survival ∼20 % - 40 % (metastatic sites, tumour burden) Schuck et al. Int J Radiat Oncol Biol Phys 2003; Womer et al. J Clin Oncol 2012; Le Deley et al J Clin Oncol 2014. 224 Initial systemic treatment • • • • Intensive chemotherapy: reported long term survival 60-70 % No novel agents available Different chemotherapy regimens standard in Europe and USA Multi-agent regimen: • • • • • vincristine (V), doxorubicin (D), actinomycin D (DTIC) cyclophosphamide (C)/ifosfamide (I) etoposide (E). Schuck et al. Int J Radiat Oncol Biol Phys 2003; Womer et al. J Clin Oncol 2012; Le Deley et al J Clin Oncol 2014. Comparison of two chemotherapy regimens in Ewing sarcoma (ES): Euro Ewing 2012 randomized trial (EE2012) • randomised study • localised or metastatic ES • patients aged 5-50 years • European regimen of VIDE induction and VAI or VAC (V, actinomycin D and I or C) consolidation • USA regimen of compressed VDC/IE induction and IE/VC consolidation Brennan et al. J Clin Oncol 2020 225 Results Brennan et al. J Clin Oncol 2020 Conclusion • The interval-compressed VDC/IE regimen is currently the preferred first-line treatment in ES • up to nine cycles of induction ChT • local therapy • consolidation chemotherapy • The optimal timing for local control: • • • • • primary site, size, response, anticipated morbidity from surgery tolerability. • Primary metastatic disease • the same treatment approach • worse prognosis • local treatment with responding metastatic disease; outcome improvement Bone sarcomas: ESMO–EURACAN–GENTURIS–ERNPaedCan: Ann Oncol 2021 226 Relapsed ES • fatal • prognostic factor: time to relapse (>2 years from initial diagnosis) • no standard of care • multiple regimens used at progression • little prospective evidence Stahl et al. Pediatr Blood Cancer 2011; Ferrari et al. Pediatr Blood Cancer 2009; Hunold et al. Pediatr Blood Cancer 2006 rEECur study • first randomised controlled trial in this setting • drug regimens: • • • • Topotecan/cyclophosphamide (TC) Irinotecan/temozolamid (TEMIRI) HD ifosfamide Gemcitabine/docetaxel (GD) McCabe et al. J Clin Oncol 2020 227 Results Irinotecan&temozolamid is less effective than topotecan/cyclophosphamide and HD ifosfamide in response rate progression free survival overall survival McCabe et al. J Clin Oncol 2020 Targeted therapy 228 Cabozanitib: CABONE study Multicentre, single arm, phase II Partial response: 26 % Stable disease: 49% Clinical benefit: 75 % Progressive disease 21% mPFS 4·4 months (95% CI 3·7–5·6) mOS 10·2 months (95% CI 8·5–18·5) *2 pts NA Cabozantinib produced high tumour shrinkage. MET expression in Ewing sarcoma (negative prognostic factor). MET inhibition might contribute to the clinical activity of cabozantinib. Italiano et al. Lancet Oncol. 2020; DuBois et al. Cancer 2010 Regorafenib: REGOBONE study • randomized, placebo -controlled phase II study • efficacy and safety of regorafenib (REG) in pts with Ewing sarcoma Regorafenib delays disease progression Only modest activity- combination therapy??? 229 Duffaud et al. ESMO 2020 Conclusion • Ewing sarcoma a rare curable cancer • Treatment in experienced reference centres • Treatment of primary tumour complex and individualised • Long term consequences of treatment very significant • Recurrence remains frequently fatal • Active new agents emerging • Testing new strategies requires coordinated international collaboration • Many improvements still needed Institute of Oncology Ljubljana a member of EURACAN Thank you for your attention! 230 2nd Summer School in Medical Oncology Institute of Oncology Ljubljana September 7-10, 2021 Presentation of Two Patients with Localised Ewing Sarcoma Aleksandra Sokolova Patient 1, December 2016: male, 27 years First visit 6/1/17 US: tumour in left hemithorax • no previous diseases • 3 weeks lump above the left lower ribs, slightly painful, growing • no other symptoms chest CT 21/12/16: 6x7 cm tumour arising from destructed anterior part of the 6th left rib, infiltrating surrounding soft tissues US guided fine needle aspiration biopsy 21/12/16 citology: Ewing sarcoma (FISH positive for EWS gene translocation) bone scintigraphy 4/1/17: no other bone lesions • examination: 6x5 cm palpable lump above lower left ribs PET/CT 5/1/17: 8 cm tumour of the 6th left rib (SUV 17.3), infiltration of surrounding soft tissues, no metastases/pathologic lymph nodes • CRP 35; LDH, AF - normal Multidisciplinary sarcoma tumour board Dec/16: male, 27 years, localised Ewing sarcoma of 6th left rib 231 January 2017 – October 2017: protocol INT-0091 neoadjuvant ChT surgery • 4 cycles VACA/IE at 3-week intervals (vincristine, doxorubicin (dactinomycin), cyclophosphamide, alternating with ifosfamide, etoposide) • after 2nd cycle the tumour no more palpable adjuvant ChT + adjuvant RT • tumour excision (Apr/2017) resection of tumour with anterior parts of 6th and 7th left ribs • histology: <5% residual Ewing sarcoma, R0 resection, narrow margins • concomitant RT to the site of the surgery (MayJul/2017, TD 50 Gy) • CT (Feb/2017): good partial regression, no new lesions Dec/16: male, 27 years, localised Ewing sarcoma of 6th left rib INT-0091: ChT + surgery + RT (Jan-Oct/17) October 2017 ⇢ today: Regular checkups (+ lab. + chest X-ray) every 3 months for the first 2 years, later every 6 months Chest CT and bone scintigraphy once a year After almost 4 years no recurrence of the disease last chest CT from 2 months ago Dec/16: male, 27 years, localised Ewing sarcoma of 6th left rib • 8 cycles VACA/IE at 3-week intervals (5th-12th cycle) • after 9th and 10th cycle febrile neutropenia gr. 4 and anemia gr. 3 • 11th and 12th cycle 75% dose INT-0091: ChT + surgery + RT (Jan-Oct/17) Follow-up Oct/17 ⇢ today: no recurrence 232 Patient 2, August 2018: male, 23 years first visit 11/9/18 • previous conditions: psoriasis • symptoms: • 9 months pain in left lumbar region and left lower ribs • progressing to both legs with tingling, difficulty walking • Aug/2018: acute paraparesis and urine retention MRI 22/8/18 9x7x6 cm left paravertebral tumour (Th11-Th12) arising from posterior part of the 12th left rib, spreading intraspinally cousing compression of medullary cone Urgent surgery 22/8/18 hemilaminectomy of Th11 and Th12 and removal of intraspinal part of the tumor -> after surgery residual mild left lumbar pain and still some weakness in legs with tingling Histology: Ewing sarcoma (fusion EWSR1-FLI1) CT thorax & abdomen and PET/CT – no distant metastases Multidisciplinary sarcoma tumour board • CRP, LDH, AF – all normal Aug/18: male, 23 years, localised Ewing sarcoma of 12th left rib September 2018 – June 2019: protocol INT-0091 neoadjuvant ChT surgery • 4 cycles VACA/IE at 3-week intervals (vincristine, doxorubicin, cyclophosphamide, dactinomycin alternating with ifosfamide, etoposide) • - after 2nd cycle complete resolution of symptoms • MRI (Oct/2018): good partial regression (9 cm -> 5 cm), no new lesions Aug/18: male, 23 years, localised Ewing sarcoma of 12th left rib adjuvant ChT + adjuvant RT • tumour excision (Dec/2018) posterior spine mobilisation with pediculotomy at Th12 and spine fixation at Th10-L1, and resection of tumour with left diaphragm and 12th rib and posterior parts of 9-11th ribs • histology: residual Ewing sarcoma with 40-50% tumour necrosis, max diameter 5 cm, R1 resection INT-0091: ChT + surgery + RT (Sep/18-Jun/19) 233 • 8 cycles VACA/IE at 3-week intervals (5th-12th cycle) • concomitant RT to the site of first and second surgery (Feb-Mar/2019, TD 58 Gy) June 2019 ⇢ regular checkups (+ lab. + chest X-ray) every 3 months, chest CT every 6 months June 2020: deterioration dyspnea, cough, right pleuritic chest pain – right pleural effusion, pneumonia; ↑CRP 200 mg/L, ↑LDH 7 ukat/L, AF norm. Chest x-ray and CT – metastases (lungs, pleura, mediastinum) June 2020: metastatic disease → 2nd line ChT: gemcitabine and docetaxel (gemcitabine day 1 and 8, docetaxel day 8, at 3-week intervals) • after 1st cycle – no more symptoms • chest X-ray after 3rd cycle: regression • total 7 cycles (Jun-Nov/2019) •PET/CT (Dec/2020): almost complete response, only one metabolically active residual mass in right upper mediastinum behind right brachiocephalic vein (2 cm, SUV 6.4) ➜ RT (Jan-Feb/2021, TD 58.8 Gy) Aug/18: male, 23 years, localised Ewing sarcoma of 12th left rib INT-0091: ChT + surgery + RT (Sep/18-Jun/19) Jun/20: metastatic 2nd line ChT (Jun-Nov/20) + RT mediastinum (Feb/21) March 2021: regular checkup Follow-up chest X-ray 15/3/2021: progression (lungs, pleura) mild stabbing pain in right chest and right shoulder, PS 0-1 CT 29/3/2021: progression – large tumour mass on right pleura infiltrating right diaphragm and liver + bone metastasis in left iliac bone 3rd line ChT: topotecan and cyclophosphamide (day 1-5 at 3-week intervals) •at initiation of therapy (Apr/2021): right chest pain spreading to right shoulder and legs, fatigue, no dyspnea or caugh; ↑CRP 200 mg/L, ↑LDH 22 ukat/L, ↑AF 3 ukat/L •after 1st cycle – disappearance of all symptoms •April/2021 ⇢ today: 6 cycles, 2-6th cycle 60% dose (3/5 days, fever on day 3) •CT (Aug/2021): good partial regression Aug/18: male, 23 years, localised Ewing sarcoma of 12th left rib INT-0091: ChT + surgery + RT (Sep/18-Jun/19) Jun/20: metastatic 2nd line ChT (Jun-Nov/20) + RT mediastinum (Feb/21) 234 3rd line ChT (Mar/21 ⇢ today) Conclusion Patient 1: male, 27 years, localised Ewing sarcoma of 6th left rib Patient 2: male, 23 years, localised Ewing sarcoma of 12th left rib Best survival with neoadjuvant ChT followed by surgery/RT if not resectable and adjuvant ChT +/- RT. 1. biopsy in sarcoma reference center 2. neoadjuvant ChT 3. surgery (R0) 4. adjuvant RT + ChT 5. follow-up – after 4 years no recurrence 1. urgent surgery due to spine compression 2. dignosis from resected part of the tumour 3. neoadjuvant ChT 4. second surgery (R1) 5. adjuvant RT + ChT 6. early recurrence 1 year after completment of treatment, currently on 3rd line of ChT All patients with radiological signs of sarcoma should be referred to the reference centre for bone sarcomas. 235 Biopsy and further treatment should be descussed at multidisciplinary sarcoma tumour board and carried out in sarcoma reference center. We should monitor cured patients for long term toxicities and secondary malignancies >10 years after treatment. Systemic treatment of prostate cancer – standards and perspectives Borislav Belev Dpt of Medical Oncology Clinic of Oncology Clinical Hospital Center Zagreb 236 237 238 239 240 241 Conclusion • 2 main trends in prostate cancer treatment: • Tendency to earlier systemic aproach • Genetic profiling being more important • New treatment options should prolong OS in prostate cancer • Personalized treatment more important and dictate optimal treatment 242 Advances in Systemic Treatment of Renal Cell Carcinoma (RCC) Boštjan Šeruga, MD, PhD Division of Medical Oncology Institute of Oncology Ljubljana and University in Ljubljana 2nd Summer School in Medical Oncology Ljubljana, September 9, 2021 Topics  Adjuvant systemic therapy in RCC ‒ Anti-VEGF agents ‒ Immune checkpoint inhibitors (ICIs)  Combined 1st line systemic therapy for advanced RCC 243 Adjuvant phase III trials in RCC 244 Adjuvant phase III trials in RCC DFS DFS DFS DFS DFS NEJM 2016 245 Adjuvant phase III trials in RCC DFS DFS DFS DFS DFS There is no role of adjuvant anti VEGF therapy Similar results in ECOG-ACRIN E2810 study, which evaluated pazopanib in this setting There is no role of anti VEGF therapy after radical metastasectomy Eur Urol Oncol 2019 246 Phase III Adjuvant Trials with Immunotherapy Parameter IMmotion010[1] (NCT03024996) PROSPER[2] (NCT03055013) Atezolizumab Drug KEYNOTE-564[3] (NCT03142334) CheckMate 914[4] (NCT03138512) Nivolumab Pembrolizumab Nivolumab + ipilimumab Histology Clear-cell ± sarcomatoid histology RCC of any histology Clear-cell ± sarcomatoid features Clear-cell ± sarcomatoid features Dose duration 1 yr 2 doses prior to surgery and adjuvant nivolumab for 9 mos 1 yr 6 mos T2 grade 4, T3a grade 3/4, T3b/c any grade, T4 any grade, or TxN+ any grade Clinical stage ≥ T2 or any N+ pT2, grade 4; pT3/4, any grade; N+ M0; M1 NED pT2aN0, grade 3-4; pT2b-T4; N+ Primary endpoint DFS RFS at 5 yrs DFS DFS BICR Yes Yes Yes Yes Active, recruiting Active, recruiting Active, recruiting Active, recruiting Risk classification Status 1. Uzzo. ASCO 2017. Abstr TPS4598. 2. Harshman. ASCO 2018. Abstr TPS4597. 3. Choueiri. ASCO 2018. Abstr TPS4599. 4. Bex. ESMO 2018. Abstr 927TiP. KEYNOTE-564: DFS (Primary Endpoint) 100 HR: 0.68 (95%: 0.35-0.87) P = .0010* DFS (%) 80 60 12-Mo Rate, % 85.7 76.2 40 24-Mo Rate, % 77.3 68.1 Pembro Placebo 20 Median follow-up: 24.1 mo (14.9-41.5) Events, % Median, Mo (95% CI) 22.0 NR (NR-NR) 30.3 NR (NR-NR) 0 0 Patients at Risk, n Pembro 496 Placebo 498 5 10 15 20 Mo 25 30 35 40 45 457 436 414 389 371 341 233 209 151 145 61 56 21 19 1 1 0 0 *Crossed P value boundary for statistical significance of .0114. Slide credit: clinicaloptions.com Choueiri. ASCO 2021. Abstr LBA5. Reproduced with permission. 247 KEYNOTE-564: DFS by Subgroup (ITT) Subgroup Events/Patients, n/N Overall Pembro Favors: Placebo HR (95% CI) 260/994 0.68 (0.53-0.87) Age  <65 yr  ≥65 yr 166/664 94/330 0.62 (0.45-0.84) 0.84 (0.56-1.26) Sex  Male  Female 79/288 181/706 0.75 (0.48-1.16) 0.66 (0.49-0.89) ECOG PS 0 1 215/847 45/147 0.65 (0.49-0.85) 0.91 (0.50-1.63) PD-L1 status  CPS <1  CPS ≥1 42/237 215/748 0.83 (0.45-1.51) 0.67 (0.51-0.88) Region  North America  European Union  Rest of world 65/258 97/375 98/361 0.87 (0.53-1.41) 0.49 (0.32-0.74) 0.81 (0.55-1.21) Metastatic staging  M0  M1 NED 234/936 26/58 0.74 (0.57-0.96) 0.29 (0.12-0.69) 0.1 0.5 1 Slide credit: clinicaloptions.com Choueiri. ASCO 2021. Abstr LBA5. Reproduced with permission. KEYNOTE-564: Interim OS 100 HR: 0.54 (95%: 0.30-0.96) P = .0164* OS (%) 80 60 12-Mo Rate, % 98.6 98.0 40 24-Mo Rate, % 96.6 93.5 Pembro Placebo 20 Median follow-up: 24.1 mo (14.9-41.5) Events, % Median, Mo (95% CI) 3.6 NR (NR-NR) 6.6 NR (NR-NR) 0 0 Patients at Risk, n Pembro 496 Placebo 498 5 10 15 20 Mo 25 30 35 40 45 490 494 486 485 482 480 338 336 215 209 124 117 51 48 3 3 0 0 *Did not cross P value boundary for statistical significance of .0000093 for 51 OS events; final OS analysis to occur after ~200 OS events. Slide credit: clinicaloptions.com Choueiri. ASCO 2021. Abstr LBA5. Reproduced with permission. 248 Take home message (adjuvant therapy)  No role of anti-VEGF adjuvant therapy in locoregional RCC after nephrectomy and after "radical" metastasectomy ‒ Conflicting results for DFS, does not improve OS, ‒ Toxic treatment, deterioration of QoL  ICIs promising new adjuvant therapy in RCC IMDC (Heng) Prognostic Criteria  Clinical Favorable Intermediate Poor ‒ KPS < 80% (P < .0001) ‒ Time from diagnosis to tx < 1 yr (P = .01)  Laboratory Probability of OS 1.0 0.8 0.6 0.4 0.2 0 ‒ Hemoglobin < LLN (P < .0001) 0 12 24 48 36 60 Mos Since Therapy Initiation ‒ Calcium > ULN (P = .0006) No. of Events/No. at Risk Favorable 11/133 Intermediate 61/301 Poor 94/152 ‒ Neutrophil count > ULN (P < .0001) 16/110 50/182 19/36 4/62 17/82 1/3 2/22 2/18 0/1 0/3 0/3 0/0 Favorable: 0 risk factors; intermediate: 1-2 risk factors; poor: 3+ risk factors ‒ Platelet count > ULN (P = .01) Heng. JCO. 2009;27:5794. 249 IMDC (Heng) Prognostic Criteria Favorable: 0 risk factors; Intermediate: 1-2 risk factors; Poor: 3+ risk factors 1.0 ‒ KPS < 80% (P < .0001) ‒ Time from diagnosis to tx < 1 yr (P = .01)  Laboratory ‒ Hemoglobin < LLN (P < .0001) Probability of OS  Clinical 0.8 Favorable 0.6 Intermediate 0.4 0.2 Poor 0 ‒ Calcium > ULN (P = .0006) 0 12 24 36 48 60 Mos Since Therapy Initiation ‒ Neutrophil count > ULN (P < .0001) ‒ Platelet count > ULN (P = .01) 75% to 80% of patients with mRCC have ≥ 1 of these risk factors (intermediate or poor risk) Heng. JCO. 2009;27:5794. Progress in the treatment of patients with advanced RCC 2-year OS of all patients (all prognostic groups) 2001-2006 IFN IFN 40% KABOZANTINIB SUNITINIB 60% 2017-2020 NIVOLUMAB + KABOZANTINIB NIVOLUMAB+CABOZANTINIB 75% 0% 10% 20% 30% 40% 50% 60% 70% 80% IFN: Interferon Choueiri T, Check Mate 9ER,ESMO 2020; Gore ME, Lancet 2010 250 Phase III clinical trials in the 1st line therapy Trial Comparison CheckMate 214 Motzer et al, NEJM, 2018 Keynote 426 Rini et al, NEJM, 2019 Javelin Renal 101 Motzer et al, NEJM, 2018 Immotion 151 Rini et al, Lancet, 2019 CheckMate 9ER Choueiri et al, ESMO 2020 Clear Motzer et al, NEJM 2021 Primary endpoint (s) Ipilimumab+Nivolumab vs. Sunitinib OS, ORR, PFS in patients with intermediate and poor prognosis Pembrolizumab+Axitinib vs. Sunitinib OS in PFS in the ITT populations (allcomers) Avelumab+Axitinib vs. Sunitinib OS in PFS in PD-L1+ patients Atezolizumab+Bevacizumab vs. Sunitinib PFS in PD-L1+ OS in the ITT population Nivolumab+Cabozantinib vs. Sunitinib PFS in the ITT population Pembrolizumab+Lenvatinib vs. Lenvatinib+Everolimus vs. Sunitinib PFS in the ITT population Results of phase III trials in the 1st line Trial CheckMate 214 Motzer et al, NEJM, 2018 Keynote 426 Rini et al, NEJM, 2019 Javelin Renal 101 Motzer et al, NEJM, 2018 Immotion 151 Rini et al, Lancet, 2019 CheckMate 9ER Choueiri et al, ESMO 2020 Clear Motzer et al, NEJM 2021 Comparison Results Ipilimumab+Nivolumab vs. Sunitinib ORR: 42% vs. 27%, PFS: 11.6 vs 8.4 mo (HR 0.82-ns), OS: at 4 yrs HR 0.65 Pembrolizumab+Axitinib vs. Sunitinib PFS: 15.1 vs. 11.1. mo (HR 0.69), at OS: 2 yrs 74% vs 66% (HR 0.68) Avelumab+Axitinib vs. Sunitinib PFS (PD-L1+): 13.8 vs 7.2 mo (HR 0.61). OS: HR 0.82 (ns-immature) Atezolizumab+Bevacizumab PFS (PD-L1+): 11.2 vs 7.7 mo (HR 0.74) vs. Sunitinib OS (ITT): HR 0.93 -ns Nivolumab+Cabozantinib vs. Sunitinib PFS (ITT): 16.6 vs. 8.3 mo (HR 0.51) OS (ITT) @ 12 mo: 85.7% vs 75.6% Pembrolizumab+Lenvatinib vs. Lenvatinib+Everolimus vs. Sunitinib PFS (ITT): 23.9 vs. 9.2 mo (HR 0.39) for P+L, OS (ITT): HR 0.66 for P+L 251 Phase III clinical trials in the 1st line therapy Trial Comparison CheckMate 214 Motzer et al, NEJM, 2018 Keynote 426 Rini et al, NEJM, 2019 Javelin Renal 101 Motzer et al, NEJM, 2018 Immotion 151 Ipilimumab+Nivolumab vs. Sunitinib CheckMate 9ER Choueiri et al, ESMO 2020 Clear Motzer et al, NEJM 2021 42% vs. 27% (9% vs 1%) Pembrolizumab+Axitinib vs. Sunitinib 59.3% vs. 35.7% (5.8% vs. 1.9%) Avelumab+Axitinib vs. Sunitinib 51.4% vs. 25.7% (3.4% vs. 1.8%) Atezolizumab+Bevacizumab vs. Sunitinib Rini et al, Lancet, 2019 ORR (CR) (%) Nivolumab+Kabozantinib vs. Sunitinib Pembrolizumab+Lenvatinib vs. Lenvatinib+Everolimus vs. Sunitinib 37% vs. 33% (5% vs. 2%) 55.7% vs. 27.1% (8% vs. 4.6%) 71% vs. 53.5% vs. 36.1% (16.8% vs. 9.8 vs. 4.2%) Long-term control of disease with ICIs Patients with intermediate and poor prognosis Checkmate 214 ≈ 70% +20% 1/3 of patients long-term control of disease Motzer RJ et al, J Immunother Cancer, 2020 252 Probability of OS OS with Nivo + Ipi vs Sunitinib in CheckMate 214: Intermediate/Poor-Risk Sarcomatoid Patients 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0 Nivo + Ipi Sun (n = 60) (n = 52) Events, n (%) 31 (52) 39 (75) Median OS, (95% CI), mos 31.2 (23.0‒NE) 13.6 (7.7‒20.9) HR (95% CI) 0.55 (0.33-0.90) P value .0155 80% (67-88) 58% (45-70) 56% (22-47) 53% (39-65) 35% (22-47) 29% (17-41) 0 3 6 9 12 15 18 27 30 33 36 39 42 45 Patients at Risk, n Nivo + ipi 60 Sun 52 21 24 Mos 56 48 52 36 49 32 47 29 45 23 43 22 37 19 30 17 29 15 22 15 10 9 5 3 1 1 0 0 32 18 Slide credit: clinicaloptions.com McDermott. KCA 2018. Which combination works earlier? Porta C, ESMO, 2020 253 KEYNOTE 426 Absolute benefits of combined therapy Subgroup 12-mo survival Pembrolizumab+ Axitinib Sunitinib Favourable 95% 94% Δ 1% Intermediate 91% 77% Δ 14% Poor 70% 45% Δ 25% ≥ 1% 90% 78% Δ 12% < 1% 92% 78% Δ 14% Prognostic group PD-L1 CPS IMDC denotes International Metastatic Renal Cell Carcinoma Database Consortium Rini et al, NEJM, 2019 Take-home Messages (advanced disease)  Combined therapy which involves ICIs is a new 1st line standard of treatment in patients with advanced RCC (for all?)  With combination therapy durable remission can be achieved also in patients with poor prognosis disease  Combination of ICI and anti VEGF preferable 1st line therapy in patients with symptomatic disease 254 255 Bladder cancer systemic treatment Is something new going on? Milena Gnjidić, UHC Zagreb, Croatia 9.2021 ~ 3% of all new cancer ~ 550 000 new cases per year worldwide ~ 200 000 death annually (2018) ~ Until the advent of immunotherapy 2015. there was no new treatment options for decades Bray F at al. GLOBOCAN. CA Cancer J Clin. 2018. 256 Cystectomy 1. Non-muscle invasive 2. Muscle invasive MULTIDISCIPLINARY TEAM 3. Metastatic Radiotherapy Bladder-sparing?! Systemic therapy Local disease Dream of every bladder-cancer patient: NO cystectomy ! Early diagnosis IMUNOTHERAPY Bladder sparing NEW THERAPY COMBINATIONS CHEMOTHERAPY RADIOTHERAPY 257 Neoadjuvant chemotherapy Cisplatin-combinations better than mono Cis ABC meta analysis. Eur Urol. 2005 Neoadjuvant chemotherapy Kim I et al. Int J Mol Sci. 2021. 258 Neoadjuvant immunotherapy Kim I et al. Int J Mol Sci. 2021. Neoadjuvant therapy Kim I et al. Int J Mol Sci. 2021. 259 Neoadjuvant therapy Kim I et al. Int J Mol Sci. 2021. Neoadjuvant therapy Could neoadj CPIs replace neoadj chemo? Combinations? New combinations? CPIs = checkpoint inhibitors Kim I et al. Int J Mol Sci. 2021. 260 Adjuvant chemotherapy Kim I et al. Int J Mol Sci. 2021. Adjuvant immunotherapy negative Kim I et al. Int J Mol Sci. 2021. 261 Adjuvant immunotherapy CheckMate 274 8.2021 FDA approved! HR 0.70 HR 0.55 DFS 20.8 ~ 10.8mo Adjuvant Nivolumab CheckMate 274 Subgroup analysis 1. PD-L1 expression > 1% ! 2. Neoadjuvant chemotherapy ! 3. Urinary bladder ! Overall survival ?! Follow-up 20 mo disease recurrence = 48.2% ~ 57.3% Bajorin DF et al. N Engl J Med. 2021. 262 Neoadjuvant vs adjuvant therapy? pro • Better tolerance • Less extensive surgery • Response monitoring • Primary tumor=more antigens • Not delaying currative cystectomy • Pathohistology CYSTECTOMY neoadjuvant adjuvant contra • Delaying cystectomy • Refractoriness to chemoth or immunoth Biomarkers !? Advanced disease Dream of every metastatic bladder-cancer patient: NO mets ! CHEMOTHERAPY Longer survival IMUNOTHERAPY NEW THERAPY COMBINATIONS 263 mBC: EAU Guidelines 2021 1 1 1 1 2 1 3 3 17 mBC: First line - chemotherapy First line Setting Regimen ORR mOS Cisplatin eligible MVAC1 40-50% 12-15 months 36-56% 7-9 months ~10% 5-8 months Gemcitabine+cisplatin2 PGC3 First line Cisplatin ineligible Gemcitabine+ Carboplatin4-6 Second line Single agent chemotherapy7-9 Single agent chemotherapy7-9 1.Loehrerer JCO 1992, 2. Van der Masse JCO 2000, 3. Bellmunt JCO 2012, 4. De Santis JCO 2012. 5. Linardou Urology 2004, 6. Nogue-Aliguer Cancer 2003, 7. Bellmunt NEJM 2017, 8.Bellmunt JCO 2009. 9. Petrilac JCO 2015. 264 mBC: First line - Chemotherpy mBC: First line - Immunotherapy only PDL1+ 50% patients cisplatin ineligible OS all ▪ Pembrolizumab ▪ Atezolizumab PDL+ Chemotherapy is still standard of care! Combinations are still pending! comb Szabados B, et al. Eur Urol Oncol. 2021. 265 mBC: First line - Maintenance JAVELIN Bladder 100 AVELUMAB OS 21.4 vs 14.3 m 9.2020 EMA approved 10.2020 21 mBC: First line - Maintenance AVELUMAB Subgroup analysis Grivas P, et al. Cancer Treat Rev. 2021. 266 22 mBC: Second line - immunotherapy EMA+ ▪ Pembrolizumab – OS 5.2021 ▪ Atezolizumab ▪ Nivolumab ▪ Avelumab ▪ Durvalumab Advantages: - longer duration - PFS - better tolerability mBC: Second line - chemotherapy First line Setting Regimen ORR mOS Cisplatin eligible MVAC1 40-50% 12-15 months Gemcitabine+cisplati n2 PGC3 First line Cisplatin ineligible Gemcitabine+ Carboplatin4-6 Second line Single agent Single agent chemotherapy7-9 chemotherapy7-9 36-56% 7-9 months ~10% 5-8 months TAXANS VINFLUNINE 1.Loehrerer JCO 1992, 2. Van der Masse JCO 2000, 3. Bellmunt JCO 2012, 4. De Santis JCO 2012. 5. Linardou Urology 2004, 6. Nogue-Aliguer Cancer 2003, 7. Bellmunt NEJM 2017, 8.Bellmunt JCO 2009. 9. Petrilac JCO 2015. 267 mBC: Second line and beyond ERDAFITINIB ( antiFGFR1-4) ENFORTUMAB VEDOTIN (ADC) Sacituzumab govitecan ( ADC) 3 Targeted therapy in mBC – today and tomorrow New therapies:         New immunotherapy New anti-FGFR New antibody-drug conjugates Anti EGFR, HER Anti VEGF New chemotherapy Vaccines CAR-T cell therapy  Combinations Nelson BE et al. Front Oncol. 2021. 268 Biomarkers Yoshida T. In J Urol. 2019. Predictive biomarkers for immunotherapy Biomarker tissue blood Predictive capacity PD-L1 Increased = improved response TMB - tumor mutational burden Increased = improved response STING – stimulator of interferon genes Deficient = reduced response ASC - apoptosis-associated speck-like protein containing CARD Gene methylation = worse survival TME - tumor microenvironment High CD4+, CD8+, CD45RO+ T cells = better survival PBMCs – peripheral blood mononuclear cells High CD4+, CD8+ T cells = better response ctDNA – ciculating tumor DNA Increased = worse survival Exsomes – extracellular vesicles Increased = better response Cytokines Predict irAEs Metal chelators Cooper chelating drugs = increase CD8+, NK sPD-L1/sPD-1 – soluble PD-L1/PD-1 Low level = better survival CTCs – circulating tumor cells Presence = poor response Microbiome Some species = better response 269 Adam T et al. Cancers. 2021 Conclusions ▪ Immunotherapy in the earlier stages is at the door ▪ Perioperative therapy is required; neoadjuvant > adjuvant ▪ Bladder-sparing approach is increasingly being on the mind ▪ Trials with new targeted therapy or combinations are ongoing ▪ Urgent need for better biomarkers to guide the optimal choice of therapy 270 Systemic treatment of germ cell tumors – could it get better BREDA ŠKRBINC INSTITUTE OF ONCOLOGY LJUBLJANA 2ND SUMMER SCHOOL IN MEDICAL ONCOLOGY 7-10 SEPTEMBER 2021 FACTS ABOUT GERM-CELL TUMORS (GCT) • GCT the most common cancer in men aged 18-35 years • Testicular cancer accounts for ~1% of newly diagnosed cancer in men worldwide Testicular cancer - NATURE REVIEWS | DISEASE PRIMERS 2018 271 FACTS ABOUT GERM-CELL TUMORS (GCT) • GCT the most common cancer in men aged 18-35 years • Testicular cancer accounts for ~1% of newly diagnosed cancer in men worldwide • Highly curable disease • Since 1975 with platinum based chemotherapy - the most curable metastatic solid cancer in males • Histology • 50%seminoma, 40% NSGCT, 10% combined GCT • Stage of the disease • 60% of patients present with lokalised disease (stage I) • Primary tumor location • Testicular, retroperitoneal, mediastinal, intracranial • Level of serum tumor markers AFP, β-hCG, LDH determine the treatment choice • IGCCCG classification PROGNOSTIC CLASSIFICATION ( y 1997 + updates) • Good prognostic group • Intermediate prognostic group • Poor prognostic group Testicular cancer - NATURE REVIEWS | DISEASE PRIMERS 2018 FACTS ABOUT GERM-CELL TUMORS (GCT) Testicular cancer - NATURE REVIEWS | DISEASE PRIMERS 2018 272 FACTS ABOUT GERM-CELL TUMORS (GCT) Multidisciplinary treatment FACTS ABOUT GERM-CELL TUMORS (GCT) • Recidivant disease still curable (probability for cure signifficantly reduced) • more efficient treatment modalities are needed for this group of GCT patients Testicular cancer - NATURE REVIEWS | DISEASE PRIMERS 2018 273 Presented By Anja Lorch at 2018 ASCO Annual Meeting Phase I / II studies Kollmansberger C trastuzumab HER2/neu expressing GCT Ann Oncol 1999 platinum refractory GCT Rick O talidomid Eur J Cancer 2006 Feldman DR sunitinib relapsed or refractory GCT Feldman DR tivantinib relapsed or refractory GCT Einhorn LH imatinibmesilat CTX refractory GCT expressing KIT J Clin Oncol 2006 Necchi A pazopanib relapsed or refractory GCT Ann Oncol 2017 Fenner M everolimus multiply relapsed GCT Adra N pembrolizumab multiply relapsed GCT, no other treatment option 274 Invest New Drugs 2010 Invest New Drugs 2013 Journal of Cancer Research and Clinical Oncology, 2018 Annals of Oncology, 2018 Le et al., Science 357, 409–413 (2017) the genomes of cancers deficient in MMR contain exceptionally high numbers of somatic mutations sensitivity to immune checkpoint blockade The overall rate of mutations per megabase in cisplatin resistan GCT is low (< 1 mutation/ MB) J Clin Oncol. 2016, 33:4000-4007. low sensitivity to immune checkpoint blokade FACTS ABOUT GERM-CELL TUMORS (GCT) • Long term survivals • risk for 2nd CA • neuropathy / hearing loss • nefropathy • risk for cardiovascular events • fertility impairment • cognitive impairment • suicidal tendencies Testicular cancer - NATURE REVIEWS | DISEASE PRIMERS 2018 275 GCT SURVIVALS How does treatment with platinum-based chemotherapy or radiotherapy influence the non–testicular cancer mortality and causes of death in GTC survivors? Investigation of cause specific non-GCT mortality with impact on previous treatment with platinum-based chemotherapy or radiotherapy The aim of the study was to assess non-TC mortality and causes of death in relation to TC treatment, including the impact of number of cisplatin based chemotherapy cycles, in a population-based cohort with complete information on TC treatment burden. 10.1200/JCO.21.00637 Journal of Clinical Oncology 2021 276 Testicular Cancer in the Cisplatin Era: Causes of Death and Mortality Rates in a Population-Based Cohort METHODS • Overall, 5,707 men identified by the Cancer Registry of Norway diagnosed with TC from 1980 to 2009 were included in this population-based cohort study. • By linking data with the Norwegian Cause of Death Registry, standardized mortality ratios (SMRs), absolute excess risks (AERs; [(observed number of deaths – expected number of deaths)/person-years of observation] x 10,000), and adjusted hazard ratios (HRs) were calculated 5707 GCT patients diagnosed with GCT Jan 1st 1980-Dec 31st 2009 identified by the Cancer Registry of Norway 10.1200/JCO.21.00637 Journal of Clinical Oncology 2021 Testicular Cancer in the Cisplatin Era: Causes of Death and Mortality Rates in a Population-Based Cohort 10.1200/JCO.21.00637 Journal of Clinical Oncology 2021 277 Testicular Cancer in the Cisplatin Era: Causes of Death and Mortality Rates in a Population-Based Cohort Results • Median follow-up was 18.7 years, during which non-TC death was registered for 665 (12%) men.  Overall excess non-TC mortality 23% (SMR, 1.23 - AER, 11.14) compared with the general population,  Increased risks after PBCT (SMR, 1.23 - AER, 7.68)  Compared with surgery, increased non-TC mortality appeared after 3, 4, and more than 4 cisplatin-based chemotherapy cycles after > 10 years of follow-up  Increased risk after RT (SMR, 1.28 - AER, 19.55). • The highest non-TC mortality was in those < 20 years at TC diagnosis (SMR, 2.27 - AER, 14.42). • The most important cause of death was non-TC second cancer with an overall SMR of 1.53 - AER, 7.94), with increased risks after PBCT and RT. • Overall noncancer mortality was increased by 15% (SMR 1.15 - AER, 4.71). • Excess suicides appeared after PBCT (SMR 1.65 - AER, 1.39). 10.1200/JCO.21.00637 Journal of Clinical Oncology 2021 Testicular Cancer in the Cisplatin Era: Causes of Death and Mortality Rates in a Population-Based Cohort Conclusions • GCT surviviors treated with platinum based CTX or RT suffer increased mortality rates compared with the general population. • The most notable excess mortality was caused by seconnd cancers, • measures to avoid delayed SC diagnosis are essential ( extende follow up) • Citotoxic treatment ( CTX / RT ) seems to be the main risk factor for increased mortality. • The increased mortality risk might be reduced by lifestyle improvements, which should be recommended following GTC treatment. • It is crucial that GCT surviviors and health personnel involved in the follow-up should be aware of the increased premature mortality risk. RISK ADAPTED TREATMENT 10.1200/JCO.21.00637 Journal of Clinical Oncology 2021 278 • Validation of the 1997 prognostic classification and update of the survival probabilities in a modern cohort • Identification of additional prognostic factors to refine the IGCCCG prognostic classification J Clin Oncol 39:1563-1574. 2021 • 30 Institutions • Complete patients database 1980-2013 J Clin Oncol 39:1563-1574. 2021 279 J Clin Oncol 39:1563-1574. 2021 J Clin Oncol 39:1563-1574. 2021 280 Nomograms of the final prognostic model for patients Without nonpulmonary visceral metastases J Clin Oncol 39:1563-1574. 2021 Nomograms of the final prognostic model for patients With nonpulmonary visceral metastases J Clin Oncol 39:1563-1574. 2021 281 https://eortc.shinyapps.io/IGCCCG-Update/ • IGCCCG database is the largest dataset on metastatic NSGCT worldwide • Improved treatment outcomes • stage migration because of earlier diagnosis and better diagnostic tools, • improved supportive care, • superiority of cisplatin- and etoposide-based first-line treatment over other combinations, • use of upfront dose-intensified regimens, • more stringent use and higher quality of postchemotherapy surgery, • better salvage strategies in nonresponding or relapsing patients, • more stringent guideline adherence, • centralization of care at experienced expert centers, • a combination of these factors J Clin Oncol 39:1563-1574. 2021 282  In future trials, patients with a particularly favorable prognosis in the nomogram may be subjected to de-escalation strategies to further reduce treatment burden in patients likely to be cured.  In contrast, trials evaluating dose-escalation strategies should be pursued in patients with the worst prognosis according to the new IGCCCG update model J Clin Oncol 39:1563-1574. 2021 The classification of the International Germ-Cell Cancer Collaborative Group (IGCCCG) has been a major advance in the management of germ-cell tumors, but relies on data of only 660 patients with seminoma treated between 1975 and 1990.  To reassess the original International Germ-Cell Cancer Collaborative Group (IGCCCG) classification for seminoma with modern treatment data and to screen for additional prognostic variables J Clin Oncol 39:1553-1562.2021 283 • 30 Institutions • Complete patients database 1980-2013 J Clin Oncol 39:1553-1562.2021 J Clin Oncol 39:1553-1562.2021 284 J Clin Oncol 39:1553-1562.2021 • LDH above 2.5 x ULN levels before chemotherapy an additional adverse prognostic factor that allowed splitting the good prognostic group further. • Good prognosis patients with an elevated LDH above 2.5 x ULN before chemotherapy experienced significantly worse outcomes. • Other variables such as age, extragonadal primary tumor, elevated levels of β-hCG, or the presence of lung metastases did not add significant prognostic information in good prognosis patients once LDH elevations were considered Until the availability of prospective trials results of this analysis do not allow to recommend using intensified CTX schemes instead of three cycles BEP in good (according to the original classification) prognosis patients who have an LDH level above 2.5 x ULN J Clin Oncol 39:1553-1562.2021 285 Given 5-year PFS and OS survival probabilities of 88% (95% CI, 87 to 89) and 95% (95% CI, 94 to 96), respectively, across all prognostic groups, metastatic seminoma represents the most curable metastatic cancer in males J Clin Oncol 39:1553-1562.2021 numerous challenges and pitfalls in testis cancer care that need to be addressed Testicular Cancer Biomarkers • Clinical management greatly relies on sTM measurements • AFP, β-hCG, LDH – specific cellular proteins secreted into the bloodstream, • conventional panel of serum tumor markers of GCT (sTMs) • standard in GCT diagnosis and treatment monitoring • Special S stage in the TNM sistem • Poor specificity and sensitivity of clasical sTM (https://doi.org/10.1155/2019/5030349) • Elevated in seminomas: bhCG 28% and LDH 29.1% , (AFP never) • Elevated in NSGCT: bhCG 53%, AFP 60.1%, and LDH 37.8% • sTMs may be elevated nonspecifically by processes other than GCT and may be normal in the setting of radiographically and serologically occult metastatic disease • Need for novel biomarkers with superior performance characteristics compared to conventional sTMs 286 https://www.nccn.org/professionals/physician_gls/pdf/testicular.pdf on horizon there is an important change in the field of GCT sTMs • In the past decade, nucleic acid–based markers, specifically microRNAs (miRNAs), have garnered attention • miRNAs - small noncoding RNA molecules of approximately 22 nucleotides • involved in epigenetic regulation of mRNA translation by direct interaction with the larger messenger RNA molecules regulating the level of gene expression on a post-transcriptional level • influencing cellular differentiation and other physiological processes as well as • carcinogenesis, for which they can operate as oncogenes or tumor suppressors Nat Rev Clin Oncol 2011;8:467–77 https://doi.org/10.1016/j.eururo.2021.06.006 287 • • Serum miRNA - the most exciting discovery in the GCT field Emerged as promising biomarkers in diagnosing and monitoring of patients with GCT • Several clusters of miRNA that are expressed in testicular cancer tissue and measurable in the serum have been identified on historical samples / retrospective studies • miR-371a-3p specifically exhibited greater accuracy than traditional sTMs in GCT https://doi.org/10.1016/j.eururo.2021.06.006 • to prospectively evaluate the utility of the M371 test in a large and representative patient population enrolled from a large number of European institutions • to involve various histologies and clinical stages and in particular • to evaluate the diagnostic sensitivity and specificity of the test for the primary diagnosis of GCT • to assess its usefulness for monitoring GCT treatment J Clin Oncol 37:1412-1423; 2019 288 J Clin Oncol 37:1412-1423; 2019 Receiver operating characteristic curves that discriminate controls from all patients with germ cell tumors, clinical stage (CS) I only or CS II/III only ROC = the measure of the usefulness of any test (a greater AUC area means a more useful test) J Clin Oncol 37:1412-1423; 2019 289 • The median expression of miR-371a-3p was significantly higher in the entire GCT group and in all the CS subgroups compared with the controls • Patients with CS greater than I had a higher serum level than those with CS I (all P < 0.001) (fig A) • Seminoma was found to have significantly lower miR-371a-3p values than nonseminoma (fig B) • difference only detectable in CS I patients • Teratoma had the lowest expression values of all subtypes (fig C) J Clin Oncol 37:1412-1423; 2019 Expression of miR-371a-3p in controls and patients with GCT recurrence. J Clin Oncol 37:1412-1423; 2019 290 Sensitivity of miR-371a-3p in all GCTs compared with the classic GCT markers b-human chorionic gonadotropin (bHCG), afetoprotein (AFP), and lactate dehydrogenase (LDH) and all three classic markers combined J Clin Oncol 37:1412-1423; 2019 Sensitivity of miR-371a-3p compared with the classic GCT markers in all GCTs CS I Sensitivity of miR-371a-3p compared with the classic GCT markers in all GCTs CS II / III Sensitivity of miR-371a-3p compared with the classic GCT markers in NSGCTs CS II / III Sensitivity of miR-371a-3p compared with the classic GCT markers in seminoma GCTs CS II / III J Clin Oncol 37:1412-1423; 2019 291 Post-treatment decrease of microRNA (miR)-371a-3p Decrease of miR-371a-3p serum levels over the course of chemotherapy in CS II patients Decrease of miR-371a-3p expression after surgical removal of the primary tumor in clinical stage (CS) I , CS II, and CS III Decreas of miR-371a-3p serum levels over the course of chemotherapy in CS III patients Decrease of miR-371a-3p expression in recurrent germ cell tumors (GCTs) after treatment J Clin Oncol 37:1412-1423; 2019 This study provides a considerable body of evidence that supports the usefulness of miR371a-3p serum levels as a new biomarker of GCTs. Five features of the M371 test are noteworthy: • the test has a 90.1% sensitivity and a 94.0% specificity for establishing the primary diagnosis of GCT; • it is relevant for the two main histologic subgroups of GCT; • miR serum levels correlate with primary tumor size, local stage, and CSs; • miR levels mirror treatment-related disease changes; • miR levels are elevated in recurrences • the study strongly confirmed previous data regarding to the usefulness of the M371 test as a new serum biomarker of GCT that is informative in both seminoma and nonseminoma; • because of its high sensitivity and specificity, M371 test involves the potential of simplifying clinical pathways of the management of GCT • further validation in an independent cohort is needed J Clin Oncol 37:1412-1423; 2019 292 The Lange-Winfild criteria for biomarkers The ideal TM is a substance that (1) is produced only by the malignancy itself; (2) is secreted into body fluids in such a way that it can easily be measured in a reproducible fashion; (3) correlates well with the amount of tumor present; (4) can be detected at an early stage of the disease; (5) has a half-life which is short enough so as not to accumulate in body tissues and result in false-positive results; (6) correlates with the response of the tumor to treatment • • M371 test has the potential of simplifying clinical pathways of the management of GCT further validation in an independent cohort is needed Cancer 60:464-472. 1987 https://doi.org/10.1016/j.eururo.2021.06.006 Systemic treatment of germ cell tumors – could it get any better? Yes it cann and it should still get better 293 Provided that • GCT patients are treated strictly according the international guidelines on diagnosis and treatment of GCT • The patients are treated by expirienced physicians dedicated to the treatment of GCT patients • GCT poor prognostic group patients are treated by multidisciplinary teams dedicated to the treatment of GCT patients 294 A new era in the treatment of endometrial cancer integrated with Molecular Classification of Endometrial Cancer-advance and reccurent disease Prof dr Aljoša Mandić President of Society for Gynecological Oncology of Serbia (UGOS) Institute for oncology of Vojvodina, Medical Faculty, University of Novi Sad, Serbia e-mail: aljosa.mandic@mf.uns.ac.rs Introduction Endometrial cancer (EC) is the most common gynecological cancer in high income countries, and the second most common in low income countries (1). In Serbia it is the fourth most common cancer in the female population – behind breast, lung and colorectal cancer (2). The molecular classification of EC introduced by the Cancer Genome Atlas (TCGA) (3) has ushered in a change in the way we view and classify EC. This project used genomic, transcriptomic, and proteomic analyses to characterize over 370 ECs. Four molecular subgroups and several predictive biomarkers have been defined based on based the genetic architecture of tumor cells. The Cancer Genome Atlas (TCGA) Research Network suggested 4 genomic classifications for endometrial cancers: a) b) c) d) polymerase ε (POLE) ultramutated, microsatellite instability hypermutated (MSI-H), copy-number low, and copy-number high. POLE ultra-mutated cancers are relatively rare (5% to 10% of endometrial cancers), highly immunogenic, and typically have a favorable prognosis.(3) MSI-H tumors have high rates of genomic mutations related to altered or defective DNA repair, including dysfunction in mismatch repair (dMMR).(3,4) Approximately 20% to 30% of endometrial cancers exhibit an MSI-H phenotype or loss of MMR. Alterations in PI3K/AKT/mTOR signaling are also common.Copy-number low tumors, or microsatellite stable (MSS) tumors, represent the majority of low-grade endometrioid cancers and have an intermediate prognosis.(3,4) These tumors have low mutation rates with few TP53 mutations, but often have mutations in PTEN, CTNNB1, PIK3CA, ARID1A, or KRAS.Copy-number high or serous-like tumors are generally high-grade with low estrogen and progesterone receptor expression and low mutation rates.(3,4) As the name suggests, these malignancies have extensive copy number variations and frequent mutation of TP53. These groups display distinct prognostic outcomes, clinical and pathological features. Although prognosis is favorable for patients with early stage disease, outcomes are poor for patients with recurrent or advanced disease. Numerous therapeutic options are available for 1 295 patients with recurrent or metastatic endometrial cancer, primarily focused on platinum-based regimens. Carboplatin in combination with paclitaxel is the option of choice (5). Recent years have witnessed the emergence of newer options for progressive disease, including bevacizumab, pembrolizumab, lenvatinib, and everolimus/letrozole. These expanded options have the potential to improve patient outcomes and provide more individualized care for patients with advanced or recurrent endometrial cancer. Patients with HER2-positive uterine serous carcinoma should be considered for the addition of trastuzumab to platinum-based doublet chemotherapy, while those with low-grade endometrioid histology may benefit from the use of hormone therapies. Targeted Therapy Antiangiogenic Therapies. In patients with advanced endometrial cancers early research indicated that there is a role for bevacizumab in certain subsets of patients, leading to phase 2 trials such as GOG-86P and MITO END-2 with the goal of testing the effectiveness of bevacizumab in combination with carboplatin/paclitaxel. (6,7). The GOG-86P trial, which also evaluated combinations with temsirolimus, did not show a significant PFS benefit for the addition of bevacizumab to platinum-doublet chemotherapy.(8). However, the ORR was 60% and median OS was significantly increased compared with historical controls from the GOG209 study (HR 0.71; P < .039). PFS was not significantly increased in any experimental arm compared to historical controls.The MITO END-2 trial failed to demonstrate a significant PFS or OS benefit for bevacizumab plus chemotherapy, although the rate of 6-month disease control was significantly higher (91% vs 70%).(7) Current NCCN Guidelines® list bevacizumab alone as an option after progression on chemotherapy or in combination with platinum-based chemotherapy for advanced disease.(5). The multi-targeted tyrosine kinase inhibitors (TKIs) lenvatinib and cediranib have also demonstrated efficacy in patients with advanced endometrial cancers.(9,10). Inhibitors of HER2 and mTOR. Uterine serous carcinomas are an aggressive variant of endometrial cancer and often have dysregulated HER2 signaling, leading to investigation of trastuzumab in combination with carboplatin/paclitaxel in this patient subset. A randomized phase 2 trial showed a 4.6-month improvement in median PFS with the addition of trastuzumab to chemotherapy (HR 0.44; P = .005) in patients with HER2-positive uterine serous carcinomas. The PFS benefit was consistent in both treatment-naive and pretreated patients, and the combination was well tolerated.(11). The phase 2 GOG-3007 trial compared hormone therapy alone with everolimus (an mTOR inhibitor) plus letrozole in patients with advanced endometrial cancer and showed an improvement in median PFS from 3.8 to 6.3 months.(12) PFS benefit from everolimus/letrozole was particularly evident in patients who had received no prior chemotherapy (median PFS: 21.6 months). This combination was well tolerated, although 2 296 rates of grade 3/4 anemia and hyperglycemia were increased compared with hormone therapy alone. Immunotherapy Several checkpoint inhibitors have demonstrated some efficacy in patients with advanced endometrial cancers.(13-20). Pembrolizumab has a tumor-agnostic approval for patients with MSI-H or dMMR advanced solid tumors and is listed in current NCCN Guidelines® as an option for advanced endometrial cancers. (5,21) Another PD-1 inhibitor, dostarlimab (TSR-042), was evaluated in the phase 2 GARNET trial, in advanced endometrial cancer. The ORR was 29.6% overall, but reached 48.8% in patients with MSI-H tumors (compared with 20.3% for those with MSS tumors). A total of 85% of patients with MSI-H endometrial cancer experienced a reduction in total tumor burden of over 50%. The DCR was 63% for MSI-H tumors vs 46.8% for those with MSS tumors.(15) Responses were durable in both cohorts, with 50% of responders remaining on therapy longer than 1 year. Dostarlimab had a manageable safety profile and was most commonly associated with fatigue, diarrhea, and nausea. Grade 3 or higher immune-mediated AEs only occurred in 5.6% of patients and included increases in alanine aminotransferase and aspartate aminotransferase, hyperglycemia, autoimmune hemolytic anemia, colitis, and infusion-related reaction. A phase 2 study evaluated avelumab in patients with dMMR and those with proficient MMR proteins.(22) The MMR-proficient cohort was closed early due to futility, while the dMMR subgroup demonstrated an ORR of 26.7%. PFS at 6 months was 40%, and responses were observed independent of PD-L1 expression. The most frequent any-grade treatment-related AEs were fatigue, nausea, hypothyroidism, neutropenia, anemia, and diarrhea. Grade 3 AEs included anemia and diarrhea, and no grade 4 toxicities were reported. A phase 1b trial recently reported data on the activity of atezolizumab in patients with advanced solid tumors, including 15 patients with recurrent or metastatic endometrial cancers. Benefit was modest, with an ORR of 13%, median PFS of 1.4 months, and median OS of 9.6 months. Benefit was more substantial in patients with high PD-L1 expression (n = 5), with an ORR of 40% and median PFS and OS of 4.2 and 38.2 months, respectively.(23) Combination regimens. Potential synergy between targeted agents and immunotherapies continues to be explored based on the hypothesis that the action of targeted therapies may prime the immune system, thus increasing the anti-immune response elicited by immunotherapies.[44] Targeted agents may enhance immunotherapy activity by improving the function or activation of immune cells, creating neoantigens, or eliciting immune cell infiltration of the tumor microenvironment. The KEYNOTE-146 trial investigated the co-inhibition of VEGF and PD-1 signaling by combining these 2 agents, lenvantinib and pembrolizumab.(25) Lenvatinib plus pembrolizumab had an ORR of 40% and a DCR of 87%. Responses were durable, with 65% having a response lasting at least 12 months. The most common any-grade AEs were fatigue, hypothyroidism, diarrhea, hypertension, decreased appetite, nausea, and stomatitis. The most frequent grade 3/4 3 297 side effects included hypertension, palmar-plantar erythrodysesthesia syndrome, and fatigue. Based on these data, lenvatinib combined with pembrolizumab recently received accelerated FDA approval for the treatment of patients with previously treated advanced endometrial carcinoma that is not MSI-H or dMMR.(21) Also concidering AE patients should be monitored regularly and educated regarding the potential for AEs to protect patient quality of life (QoL) and avoid unnecessary treatment delays or interruptions. Some others ongoing clinical trials continue to investigate immunotherapeutic combination approaches for endometrial cancer, including combinations with chemotherapy such as phase 2 trial is evaluating pembrolizumab with carboplatin/paclitaxel in patients with advanced endometrial carcinoma.(26) The phase 3 RUBY trial is directly comparing carboplatin/paclitaxel with placebo or dostarlimab in patients with recurrent or primary advanced endometrial cancer.(27) The phase 3 AtTEnd trial is evaluating atezolizumab plus carboplatin/paclitaxel in patients with advanced or recurrent endometrial cancer.(28) Another area of interest is the potential efficacy of combining immune checkpoint inhibitors with inhibitors of poly(ADP-ribose) polymerase (PARP). Ongoing phase 2 trials are investigating combinations such as olaparib/durvalumab (DOMEC trial), rucaparib/atezolizumab/bevacizumab (EndoBARR trial), and niraparib/dostarlimab in patients with recurrent or metastatic endometrial cancers.(29-31) Highlights Given the molecular subtypes of endometrial cancer outlined by TCGA, optimal patient selection for emerging therapies is going to be paramount to ensuring patients receive the most effective therapy without compromising safety and QoL. Despite randomized phase II trials suggesting that adding bevacizumab to chemotherapy carboplatin and paclitaxel in frontline endometrial cancer was feasible and may result in some benefit, no randomized phase III trials have been conducted to confirm those results Immune checkpoint inhibitors have shown efficacy in patients with recurrent MSI-H/dMMR solid tumors, including endometrial cancers, with a prolonged duration of response in those who respond Single-agent immunotherapy with anti–PD-1 monoclonal antibodies, either dostarlimab or pembrolizumab, is approved for patients with recurrent endometrial cancer with MSI-H/dMMR. Still, experts suggest that adding lenvatinib to pembrolizumab monotherapy in patients with MSI-H tumors is likely to result in added toxicity, but not meaningfully improve response Response with single-agent pembrolizumab in MSI-H endometrial cancer in KEYNOTE-158 is comparable to that of the combination of lenvatinib plus pembrolizumab in MSI-H/dMMR endometrial cancers from KEYNOTE-146 (57.1% vs 63.6%) Single-agent checkpoint inhibitor is the preferred choice in MSI-H/dMMR endometrial cancer 4 298 Also have to notice that many of the currently available therapies are associated with AEs that can negatively affect patient QoL and lead to early treatment discontinuation. Unique targets continue to be identified, and ongoing clinical trials will hopefully elucidate promising therapies to improve survival and QoL for patients with advanced disease. References: 1. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries 2. World Health Organization International Agency for Research on Cancer (IARC). GLOBOCAN 2020: Corpus uteri fact sheet. [homepage on the internet]; 2020 [cited 2032 Apr 23]. Available from: https://gco.iarc.fr/today/data/factsheets/cancers/24-Corpusuteri-fact-sheet.pdf 3. Cancer Genome Atlas Research Network, Kandoth C, Schultz N, et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013; 497: 67-73. 4. Di Tucci C, Capone C, Galati G, et al. Immunotherapy in endometrial cancer: new scenarios on the horizon. J Gynecol Oncol. 2019;30:e46. 5. National Comprehensive Cancer Network. Clinical practice guidelines in oncology. Uterine neoplasms. Version 5.2019. https://www.nccn.org/professionals/physician_gls/pdf/uterine.pdf. Accessed February 10, 2020. 6. Aghajanian C, Sill MW, Darcy KM, et al. Phase II trial of bevacizumab in recurrent or persistent endometrial cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2011;29:2259-2265. 7. Lorusso D, Ferrandina G, Colombo N, et al. Carboplatin-paclitaxel compared to carboplatin-paclitaxel-bevacizumab in advanced or recurrent endometrial cancer: MITO END-2 - a randomized phase II trial. Gynecol Oncol. 2019;155:406-412. 8. Aghajanian C, Filiaci V, Dizon DS, et al. A phase II study of frontline paclitaxel/carboplatin/bevacizumab, paclitaxel/carboplatin/temsirolimus, or ixabepilone/carboplatin/bevacizumab in advanced/recurrent endometrial cancer. Gynecol Oncol. 2018;150:274-281. 9. Vergote I, Powell MA, Teneriello MG, et al. Second-line lenvatinib in patients with recurrent endometrial cancer. Gynecol Oncol. 2020. doi: 10.1016/j.ygyno.2019.12.039. [Epub ahead of print] 10. Bender D, Sill MW, Lankes HA, et al. A phase II evaluation of cediranib in the treatment of recurrent or persistent endometrial cancer: an NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol. 2015;138:507-512. 11. Fader AN, Roque DM, Siegel E, et al. Randomized phase II trial of carboplatin-paclitaxel versus carboplatin-paclitaxel-trastuzumab in uterine serous carcinomas that overexpress human epidermal growth factor receptor 2/neu. J Clin Oncol. 2018;36:2044-2051. 12. Slomovitz BM, Filiacic VL, Coleman RL, et al. GOG 3007, a randomized phase II (RP2) trial of everolimus and letrozole (EL) or hormonal therapy (medroxyprogesterone 5 299 acetate/tamoxifen, PT) in women with advanced, persistent or recurrent endometrial carcinoma (EC): a GOG Foundation study. Presented at: 49th Annual Meeting of the Society of Gynecologic Oncology; March 24-27, 2018; New Orleans, LA. Abstract 1. 13. Ott PA, Bang YJ, Berton-Riqaud D, et al. Safety and antitumor activity of pembrolizumab in advanced programmed death ligand 1-positive endometrial cancer: results from the KEYNOTE-028 study. J Clin Oncol. 2017;35:2535-2541. 14. Marabelle A, Le DT, Ascierto PA, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38:1-10. 15. Oaknin A, Duska LR, Sullivan RJ, et al. Preliminary safety, efficacy, and pharmacokinetic/pharmacodynamic characterization from GARNET, a phase I/II clinical trial of the anti-PD-1 monoclonal antibody, TSR-042, in patients with recurrent or advanced MSI-H and MSS endometrial cancer. Presented at: 50th Annual Meeting of the Society of Gynecologic Oncology; March 16-19, 2019; Honolulu, HI. Abstract 33. 16. Azad NS, Gray RJ, Overman MJ, et al. Nivolumab is effective in mismatch repairdeficient noncolorectal cancers: results from arm Z1D-A subprotocol of the NCIMATCH (EAY131) study. J Clin Oncol. 2020;38:214-222. 17. Konstantinopoulos PA, Luo W, Liu JF, et al. Phase II study of avelumab in patients with mismatch repair deficient and mismatch repair proficient recurrent/persistent endometrial cancer. J Clin Oncol. 2019;37:2786-2794. 18. Antill YC, Kok PS, Robledo K, et al. Activity of durvalumab in advanced endometrial cancer (AEC) according to mismatch repair (MMR) status: the phase II PHAEDRA trial (ANZGOG1601). J Clin Oncol. 2019;37(suppl): Abstract 5501. 19. Fleming GF, Emens LA, Eder JP, et al. Clinical activity, safety and biomarker results from a phase Ia study of atezolizumab (atezo) in advanced/recurrent endometrial cancer (rEC). J Clin Oncol. 2017;35(suppl): Abstract 5585. 20. Makker V, Rasco D, Vogelzang NJ, et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: an interim analysis of a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019;20:711-718.44. 21. KEYTRUDA (pembrolizumab) [prescribing information]. Whitehouse Station, NJ: Merck & Co., Inc.; Approved 2014. Revised January 2020. 22. Konstantinopoulos PA, Luo W, Liu JF, et al. Phase II study of avelumab in patients with mismatch repair deficient and mismatch repair proficient recurrent/persistent endometrial cancer. J Clin Oncol. 2019;37:2786-2794. 23. Fleming GF, Emens LA, Eder JP, et al. Clinical activity, safety and biomarker results from a phase Ia study of atezolizumab (atezo) in advanced/recurrent endometrial cancer (rEC). J Clin Oncol. 2017;35(suppl): Abstract 5585. 24. Hughes PE, Caenepeel S, Wu LC. Targeted therapy and checkpoint immunotherapy combinations for the treatment of cancer. Trends Immunol. 2016;37:462-476. 25. Makker V, Rasco D, Vogelzang NJ, et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: an interim analysis of a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019;20:711-718.44. 6 300 Pembro/carbo/taxol in endometrial cancer. 26. Clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT02549209. Accessed February 26, 2020. 27. Clinicaltrials.gov. A study of dostarlimab (TSR-042) plus carboplatin-paclitaxel versus placebo plus carboplatin-paclitaxel in patients with recurrent or primary advanced endometrial cancer (RUBY). https://clinicaltrials.gov/ct2/show/NCT03981796. Accessed February 26, 2020. 28. Clinicaltrials.gov. Atezolizumab trial in endometrial cancer - AtTEnd (AtTEnd). https://clinicaltrials.gov/ct2/show/NCT03603184. Accessed February 26, 2020. 29. Clinicaltrials.gov. Durvalumab and olaparib in metastatic or recurrent endometrial cancer (DOMEC). https://clinicaltrials.gov/ct2/show/NCT03951415. Accessed February 27, 2020. 30. Clinicaltrials.gov. The EndoBARR trial (Endometrial Bevacizumab, Atezolizumab, Rucaparib) (EndoBARR). https://clinicaltrials.gov/ct2/show/NCT03694262. Accessed February 27, 2020. 31. Clinicaltrials.gov. Study of niraparib and TSR-042 in recurrent endometrial cancer. https://clinicaltrials.gov/ct2/show/NCT03016338. Accessed February 27, 2020. 7 301 The Early and locally advanced hormone dependant Breast cancer – where do we stand? Semir Beslija Oncology Division KCUS EARLY BREAST CANCER: WHO NEEDS ADJUVANT ET? • (almost) All ER+ EARLY BREAST CANCER patients! 302 Identifying Patients at Biological High Risk for Recurrence Genomic features Proliferation features  Gene expression assays  Ki-67 by immunohistochemistry – 21-gene (Oncotype DX) – 70-gene (MammaPrint) – Cutoff values of High vs Low WSG PlanB Trial: DFS by Oncotype RS and Ki-67 DFS in HR+/Ki-67 >10% and <40% 80 60 40 RS Group RS 0-11 20 RS 12-25 RS >25 0 0 12 24 36 Mo 48 60 DFS by Ki-67 Expression Levels: HR+ 100 Disease-Free Survival Disease-Free Survival 100 PlanB: DFS by Ki-67[a] 80 60 40 HR+/Ki-67 0-10% HR+/Ki-67 >10% - <40% HR+/Ki-67 ≥40% HR-/HER2- 20 0 72 0 12 24 36 Mo 48 60 72 Nitz. Breast Cancer Res Treat. 2017;165:573. TAILORx: Treatment Assignment and Randomization  Randomized, parallel-assignment phase III study Patients with node-negative HR+, HER2-, breast cancer T1c-T2 (high-risk T1b) (N = 10,273) Stratified by RS (11-15, 16-20, 2125), menopausal status (pre vs post), planned chemotherapy (taxane vs not), and planned radiation Baseline biopsy evaluated for RS (Oncotype DX) RS ≤10 Arm A: ET Alone (n = 1629) Arm B: ET Alone (n = 3399) RS 11-25 Arm C: ET + Chemotherapy (n = 3312) 9719 patients randomized RS ≥26 Statistical Design Noninferiority - iDFS Hazard Ratio: 1.332 (90 vs. 87% 5-yr DFS) Type I 10%, type II 5% Full info: 835 iDFS events Arm D: ET + Chemotherapy (n = 1389)  Primary endpoint: iDFS, secondary primary cancer, or death  Key secondary endpoints: freedom from breast cancer recurrence at distant site, freedom from breast cancer recurrence at distant or local-regional site, OS NCT00310180. Sparano. NEJM. 2018;379:111 303 TAILORx: ITT Population—RS 11-25 (Arms B & C)  836 iDFS events (after median of 7.5 yr), including 338 (40.4%) with recurrence as first event, of which 199 (23.8%) were distant DFS Probability 1.0 Distant Recurrence-Free Probability Primary Endpoint Invasive Disease–Free Survival 0.8 P =.26 Hazard ratio, ET alone vs CT + ET: 0.6 1.08 (95% CI: 0.94-1.24) 0.4 0.2 0 Patients at 0 Risk, n CT + ET 3312 ET alone 3399 CT + ET ET alone 12 24 36 48 60 Mo 72 84 96 108 3204 3293 3104 3194 2993 3081 2849 2953 2335 2431 1781 1859 1130 1197 2645 2741 523 537 Secondary Endpoint Distant Relapse-Free Interval 1.0 0.8 P =.48 Hazard ratio, ET alone vs CT + ET: 0.6 1.10 (95% CI: 0.85-1.41) 0.4 0.2 0 Patients at 0 Risk, n CT + ET 3312 ET alone 3399 CT + ET ET alone 5 12 24 36 48 60 Mo 72 84 96 108 3215 3318 3142 3239 3059 3147 2935 3033 2432 2537 1866 1947 1197 1267 2734 2833 554 581 Sparano. NEJM. 2018;379:111. RxPONDER: Adjuvant ET ± Chemotherapy in HR+/HER2EBC With 1-3 Positive Lymph Nodes and RS ≤25  Randomized phase III trial Stratified by RS (0-13 vs 14-25), menopausal status (pre vs post), axillary surgery (ALND vs SLNB) Adults with HR+/HER2- EBC and 1-3 positive LN without distant mets*; able to receive adjuvant taxane and/or anthracycline-based CT†; axillary staging by SLNB or ALND; RS 0-25‡ (N = 5015) Chemotherapy Followed by ET (n = 2509) ET Alone (n = 2506) Baseline characteristics generally well balanced between treatment arms *Protocol amended to exclude patients with pN1mic as only nodal disease after 2493 patients randomized. †Approved CT regimens: TC, FAC (or FEC), AC/T (or EC/T), FAC/T (or FEC/T); AC alone or CMF not allowed. ‡Patients with RS > 25 recommended to be treated with CT followed ET off study.  Primary endpoint: iDFS  Key secondary endpoints: OS, distant DFS, local DFI, toxicity, QoL Kalinsky. SABCS 2020. Abstr GS3-00. 304 RxPONDER: iDFS (Primary Endpoint)  In this population of node positive, hormone receptor–positive BC with RS 0-25, RS did not predict relative CT benefit for iDFS iDFS in Overall Population iDFS Probability 1.0 ‒ HR: 1.02 (95% CI: 0.98-1.06; P = .30)  CT use and RS independently prognostic for iDFS ‒ iDFS events less likely among patients who received CT 0.8 CT + ET (n = 2509) 0.6 Events 198 249 5-yr iDFS, % 92.4 91.0 0.4 0.2 0 ET (n = 2506) Absolute difference, % 1.4 HR: 0.81 (95% CI: 0.67-0.98; P = .026) 0 1 2 3 4 5 6 7 Patients at Yr Since Randomization Risk, n CT + ET 2509 2277 2104 1893 1648 1397 857 403 ET 2506 2327 2161 1910 1696 1404 846 397 ‒ HR: 0.81 (95% CI: 0.67-0.96; P = .026) ‒ iDFS events more likely among patients with higher RS 8 9 122 135 4 11  At median follow-up of 5.1 yr, 447 iDFS events were observed (54% of expected at final analysis) ‒ HR: 1.06 (95% CI: 1.04-1.07; P <.001) Kalinsky. SABCS 2020. Abstr GS3-00. RxPONDER: iDFS by Menopausal Status Postmenopausal Premenopausal 1.0 0.8 CT + ET (n = 1675) 0.6 0.2 0 ET (n = 1675) Events 147 158 5-yr iDFS, % 91.6 91.9 0.4 Absolute difference, % iDFS Probability iDFS Probability 1.0 NS HR: 0.97 (95% CI: 0.78-1.22; P = .82) 0 1 2 3 4 5 6 7 Yr Since Randomization Patients at Risk, n CT + ET 1675 1514 1400 1268 1113 ET 1675 1567 1462 1308 1167 943 975 585 601 287 298 8 9 88 104 3 9 0.8 CT + ET (n = 834) 0.6 Events 0.4 5-yr iDFS, % 0.2 0 ET (n = 831) 51 91 94.2 89.0 Absolute difference, % 5.2 HR: 0.54 (95% CI: 0.38-0.76; P = .0004) 0 Patients at Risk, n CT + ET 834 ET 831 1 2 3 4 5 6 7 Yr Since Randomization 763 760 704 699 625 602 535 529 454 429 272 245 116 99 8 9 34 31 1 2  Absolute difference in distant recurrence as first site: 2.9% (3.1% CT + ET vs 6.0% ET)  Absolute difference in distant recurrence as first site: 0.3% (2.3% CT + ET vs 2.6% ET) Kalinsky. SABCS 2020. Abstr GS3-00. 305 Chemotherapy for ER+/HER2- BC With 0-3 LN RS 0-10 RS 11-15 RS 16-20 RS 21-25 RS ≥26 Postmenopausal LN positive LN negative Premenopausal LN positive ▲ 6.2% (RS 14-25) ▲ 3.9% (RS 0-13) LN negative Low Clin Risk ▲ 0.2% ▲ 6.4% High Clin Risk ▲ 6.5% ▲ 8.7% ET alone CT (discuss OFS + AI as alternative) CT + ET ▲ Denotes benefit (dDFS for LN- and iDFS for LN+) with addition of CT Kalinsky. SABCS 2020. Abstr GS3-00. Sparano. NEJM. 2019;380:2395. ADAPT HR+/HER2-: Adjuvant ET ± Chemotherapy in Intermediate/High-Risk, HR+/HER2- Luminal EBC  2-part, prospective phase III trial ‒ Part 1: Current analysis evaluated prognostic impact of RS <26 and Ki-67 decrease after shortcourse of preoperative ET in the ET alone arm and is not a randomized comparison Adult patients with HR+/HER2unilateral luminal EBC; cT1-4c, cN0-3; candidates for adjuvant CT by conventional prognostic criteria* (N = 4691) Baseline biopsy evaluated for RS (Oncotype DX) and Ki-67 expression; surgical specimen evaluated for Ki-67 expression† after short ET run-in *cT2 or G3 or Ki-67 ≥15% or <35 yr old or cN+. †Ki-67 post ≤10% = ET response.  Primary endpoint: 5-yr iDFS ‒ Part 1: NI* for pN0-1/RS 12-25/Ki-67post ≤10% vs pN0-1/RS 0-11 Harbeck. SABCS 2020. Abstr GS4-04.  pN2-3  pN0-1/RS >25  pN0-1/RS 12-25/ Ki-67post > 10% CT Followed by ET (n = 2335)  pN0-1/RS 12-25/ Ki-67post ≤10%  pN0-1/RS 0-11 ET Alone (n = 2356)  Key secondary endpoints: dDFS, OS, translational research *NI defined as ≤3.3% 1-sided 95% CL of 5-yr iDFS difference (α = .05, 80% power; 5% dropouts) 306 ADAPT HR+/HER2-: 5-Yr iDFS (Primary Endpoint)  Primary endpoint met 100 ─ 5-yr iDFS difference: -1.3% (95% CI: -3.3% to 0.6%) iDFS (%) 80 5-Yr iDFS, % (95% CI) RS 0-11 93.9 (91.8-95.4) RS 12-25/Ki-67 ≤10% 92.6 (90.8-94.0) 60 40 ─ 95% lower confidence limit of -3.3% met prespecified criterion for noninferiority of pN0-1/RS 12-25/ Ki-67post ≤10% vs pN0-1/RS 0-11 (P = .05) 20 0 0 Patients at Risk, n RS 0-11 865 RS 12-25/Ki-67 ≤10% 1414 12 796 1289 24 36 Follow-up (Mo) 705 1124 657 1019 48 60 603 938 431 671  5-yr OS rate ─ 97.3% for pN0-1/RS 12-25/Ki-67 ≤10% vs 98.0% for pN0-1/RS 0-11 (P = .160) Harbeck. SABCS 2020. Abstr GS4-04. Treatment De-Escalation Strategies in HR+/HER2- EBC: Summary TAILOR-x :In LN-negative breast cancer:  ‒ Age >50 yr: RS ≤25 have no chemotherapy benefit ‒ Age ≤50 yr: RS 16-25 may derive chemotherapy benefit RxPONDER: analysis of adj CT for HR+/HER2- EBC with 1-3 positive nodes and RS ≤25, postmenopausal women did not benefit, whereas premenopausal women did1  ‒ Premenopausal patients experienced a 46% decrease in iDFS events and a 53% decrease in deaths, leading to a 5-yr OS absolute improvement of 1.3%  ADAPT HR+/HER2-: primary endpoint reached: patients with luminal EBC and 0-3 positive nodes, RS 12-25 and endocrine response (Ki-67post ≤10%)2 after short preoperative ET had a 5-yr iDFS (92.6%) comparable to those with 0-3 LN and RS 0-11 (93.9%)  Outcome was excellent in both groups with adj ET alone: 5-yr dDFS (95.6% vs 96.3%) and 5-yr OS (97.3% vs 98.0%) 1. Kalinsky. SABCS 2020. Abstr GS3-00. 2. Harbeck. SABCS 2020. Abstr GS4-04. 307 Is There a Role for CDK4/6 Inhibition for Early-Stage HR+ Disease? PENELOPE-B palbociclib Estimated Annual Hazard Rate Risk of First Recurrence After Primary Treatment 0.25 Stage I II After neoadjuvant, high risk III 0.20 monarchE 0.15 abemaciclib High-risk CPR factors, Ki-67 0.10 PALLAS palbociclib 0.05 Stage II, III 0 0 2 4 6 8 10 Follow-up Time After Primary Diagnosis of Breast Cancer (Yr) NATALEE ribociclib Stage II, III Cheng. Cancer Epidemiol Biomarkers Prev. 2012;21:800. PALLAS: Phase III Open-Label Study of Adjuvant Palbociclib + Endocrine Therapy  Multicenter, open-label, randomized phase III trial Stratified by stage (IIA vs IIB/III), chemotherapy (yes vs no), age (≤50 vs >50 yr), geographic region (N America vs Europe vs other) Palbociclib x 2 yr*+ Endocrine therapy† (n = 2883) Patients with stage II-III HR+/HER2breast cancer; completion of prior surgery, ± CT, RT within 12 mo of diagnosis or within 6 mo of starting adjuvant endocrine treatment; FFPE tumor block submitted (N = 5760) Endocrine therapy† (n = 2877) *125 mg QD, 3 wk on/1 wk off †Aromatase inhibitor or tamoxifen ± LHRH agonist.  Primary endpoint: invasive disease–free survival Mayer. ESMO 2020. Abstr LBA12. Mayer. Lancet Oncol. 2021;22:212. 308 PALLAS: Primary Endpoint iDFS iDFS 100 100 90 90 80 80 60 50 40 30 20 iDFS Palbociclib + ET ET Alone 88.2% 88.5% 70 DRFS (%) 70 iDFS (%) DRFS Hazard ratio: 0.93 (95% CI: 0.76-1.15; P = .51) Arm Events Palbociclib + ET 170 ET 181 10 P = .51 Hazard ratio: 0.93 (95% Cl: 0.76-1.15) 0 0 6 12 18 60 50 20 0 36 90.7% P = .9997 Hazard ratio: 1.00 (95% Cl: 0.79-1.27) 10 30 ET Alone 89.3% Arm Events Palbociclib + ET 136 135 ET 30 24 Palbociclib + ET Hazard ratio: 1.00 (95% CI 0.79-1.27; P = .9997 40 Mo From Randomization  DRFS 0 6 12 18 24 30 Mo From Randomization At a median follow-up of 23.7 mo, no significant difference in either 3-yr iDFS or DRFS was observed Mayer. ESMO 2020. Abstr LBA12. Mayer. Lancet Oncol. 2021; 22:212. monarchE: Adjuvant Abemaciclib + ET in High-Risk, Node-Positive, HR+/HER2- EBC  International, randomized, open-label phase III trial ITT Population (Cohorts 1 + 2) Women or men with high-risk, node-positive, HR+/HER2- EBC; prior (neo)adjuvant CT permitted; pre- or postmenopausal no distant metastasis; ≤16 mo from surgery to randomization; ≤12 wk of ET after last non-ET (N = 5637) Stratified by prior CT, menopausal status, region Cohort 1 ≥4 positive ALN or 1-3 positive ALN plus histologic grade 3 and/or tumor ≥5 cm Abemaciclib 150 mg BID up to 2 yr + ET per standard of care of physician’s choice for 5-10 yr as clinically indicated (n = 2808) Cohort 2 1-3 positive ALN, Ki-67 ≥20% per central testing, not grade 3, tumor size <5 cm ET per standard of care of physician’s choice for 5-10 yr as clinically indicated (n = 2829)  Primary endpoint: iDFS ‒ Planned for after ~390 iDFS events (~85% power, assumed iDFS hazard ratio of 0.73, cumulative 2-sided α = 0.05) ‒ Current primary outcome efficacy analysis occurred after 395 iDFS events in ITT population  Key secondary endpoints: iDFS in Ki-67 high (≥20%) population, distant RFS, OS, safety, PRO, PK Johnston. JCO. 2020;38:3987. Rastogi. SABCS 2020. Abstr GS1-01. 309 36 iDFS (%) monarchE: iDFS (Primary Endpoint) 100 90 80 70 60 50 40 30 20 10 0 Median f/u: 19.1 mo in both arms. Curves should not be interpreted beyond 24 mo due to limited f/u. 0 Patients at Risk, n Abe + ET 2808 ET 2829 Abemaciclib + ET (n = 2808) ET (n = 2829) Events, n 163 232 2-yr iDFS, % 92.3 89.3 Hazard ratio: 0.713 (95% CI: 0.583-0.871; P = .0009) 3 6 9 12 15 2680 2700 2619 2653 2573 2609 2519 2548 2076 2093 18 21 24 27 30 33 36 1487 1499 1029 1033 619 627 133 131 94 102 1 0 0 0 Mo Johnston. JCO. 2020;38:3987. Rastogi. SABCS 2020. Abstr GS1-01. Why Did MonarchE Succeed Where PALLAS Failed? MonarchE iDFS PALLAS iDFS 100 90 iDFS (%) 80 70 60 Palbociclib + ET ET Alone 88.2 88.5 3-yr iDFS, % 50 HR: 0.93 (95% CI: 0.76-1.15; log-rank P = .51) 40 Event, n Palbociclib + ET 170 ET 181 30 20 10 Log-rank P = .51 0 0 Patients at Risk, n Palbociclib + ET 2883 ET 2877 6 2634 2649 12 18 24 30 36 2563 2535 1946 1953 1257 1295 583 574 163 172 Mo From Randomization Differences between the drugs themselves/drug exposure and discontinuations/ level of risk within patient populations? Johnston. JCO. 2020;38:3987. Mayer. ESMO 2020. Abstr LBA12. Mayer. Lancet Oncol. 2021;22:212. 310 monarchE NAC Subgroup Analysis: Prior NAC Regimen Prior NAC Regimen, n (%) Abemaciclib + ET (n = 1025) ET Alone (n = 1031) Anthracycline + taxane 903 (88.1) 931 (90.3) Anthracycline without taxane 71 (6.9) 59 (5.7) Taxane + cyclophosphamide 28 (2.7) 23 (2.2) Other* 23 (2.2) 18 (1.7)  NAC subgroup comprised 36% of ITT population  Most patients received standard NAC regimen of anthracycline + taxane ‒ Use of standard NAC regimen was consistent between treatment arms *Includes taxane only, cyclophosphamide only, other CT. Martin. ASCO 2021. Abstr 517. monarchE NAC Subgroup Analysis: iDFS (Primary Endpoint) 100 iDFS (%) 80 NAC Subgroup 60 Abemaciclib + ET ET Alone 92 148 iDFS events, n 40 HR (95% CI) 20 0 0.614 (0.473-0.797; nominal P = .0002) 2-yr iDFS rate, % 87.2 Abemaciclib + ET ET alone ITT Population Abemaciclib + ET ET Alone -- -- 0.713 (0.583-0.871) 80.6 -- -- Difference: 6.6% 0 Patients at Risk, n Abemaciclib + ET 1025 ET alone 1031 3 6 9 12 15 976 971 948 948 922 923 904 891 728 717  In the NAC subgroup, abemaciclib + ET demonstrated a clinically meaningful 38.6% reduction in risk of an iDFS event vs ET alone 18 Mos 500 499 21 24 27 30 33 36 347 334 203 194 43 33 29 23 1 0 0 0  The 2-yr iDFS rate was higher with abemaciclib + ET vs ET alone in the NAC subgroup (87.2% vs 80.6%; difference: 6.6%) Martin. ASCO 2021. Abstr 517. Reproduced with permission. 311 monarchE NAC Subgroup Analysis: Conclusions  In this preplanned subgroup analysis of the monarchE trial, abemaciclib + adjuvant ET demonstrated clinically meaningful improvements in iDFS and distant RFS vs ET alone in patients with high-risk HR+/HER2- EBC who received prior NAC1 ‒ Reduction in risk: iDFS, 38.6%; distant RFS, 39.1% ‒ Benefits were numerically greater than those observed in ITT population and were maintained independent of tumor size at diagnosis and surgery  Among those treated with ET alone, the NAC subgroup exhibited a lower 2-yr iDFS rate vs the ITT population consistent with a higher risk of recurrence1-3 ‒ 2-yr iDFS rate comparable to that reported in control arm of phase III PENELOPE-B trial, which compared palbociclib + ET vs placebo + ET in women with high-risk HR+/HER2- EBC after NAC4  Safety profile in this population consistent with prior reports for abemaciclib1 21 1. Martin. ASCO 2021. Abstr 517. 2. Rastogi. SABCS 2020. Abstr GS1-01. 3. Johnston. JCO. 2020;38:3987. 4. Loibl. JCO. 2021;39:1518. PENELOPE-B: Palbociclib + ET in HR+/HER2- BC at High Risk of Relapse After Neoadjuvant Chemotherapy  Randomized, double-blind, placebo-controlled phase III trial Stratified by age (≤50 vs >50 yr), nodal status (ypN0-1 vs ypN2-3), Ki-67 (>15% vs ≤15%), region (Asia vs non-Asia), and CPS-EG score (≤3 vs 2 and ypN+) Adult patients with confirmed HR+/HER2- BC with residual disease after ≥16 wk of neoadjuvant CT*; CPS-EG score ≥3 or 2 with ypN+ (N = 1250) Palbociclib 125 mg QD Days 1-21 28-day cycles x 13 + ET by local standard (n = 631) Surgery ± radiotherapy† Placebo QD Days 1-21 28-day cycles x 13 + + ET by local standard (n = 619) *Includes 6 wk of taxanes. †Time between locoregional therapy and randomization: <16 wk from final surgery or <10 wk from RT completion.  Primary endpoint: iDFS  Secondary endpoints include: iDFS excluding second primary invasive non-breast cancers, distant DFS, locoregional RFS, OS, safety, compliance, QoL Loibl. JCO 2021;10:1200. 312 PENELOPE-B: iDFS (Primary Endpoint) 2-Yr iDFS 88.3% 3-Yr iDFS 81.2% 4-Yr iDFS 73.0% 84.0% 77.7% 72.4% 100 iDFS (%) 80 60  Median f/u: 42.8 mo  Types of iDFS events – 74% distant recurrences – 116 with palbociclib, 111 with placebo iDFS Events, n Palbociclib + ET 152 Placebo + ET 156 40 – 16% invasive locoregional recurrences Hazard ratio: 0.93 (95% CI: 0.74-1.17; P = .525) 20 – 21 with palbociclib, 27 with placebo + Censored 0 Patients at 0 Risk, n Palbociclib 631 Placebo 619 12 571 553 24 36 48 60 72 528 497 Mo 389 349 169 161 38 24 0 1 Loibl. JCO 2021;10:1200. CDK4/6 Inhibition in High-Risk HR+/HER2- EBC: Summary  monarchE: In a preplanned interim analysis, adj abemaciclib + ET continued to demonstrate improved iDFS vs ET alone for HR+/HER2- EBC at high risk of relapse after locoregional tx and/or (neo)adj CT (hazard ratio: 0.75; 95% CI: 0.60-0.93; P = .01)1,2 ‒ 2-yr iDFS rates: 92.2% with abemaciclib + ET vs 88.7% with ET ‒ Significant iDFS improvement observed in Ki-67 high (≥20%) tumors ‒ Distant RFS also improved (hazard ratio: 0.72; 95% CI: 0.56-0.92; P = .01), with 2-yr distant RFS rates of 93.6% vs 90.3%, respectively  PENELOPE-B: In the first interim analysis, the addition of 1 yr of adjuvant palbociclib to ET in the curative setting failed to demonstrate a benefit in patients with higher-risk HR+/HER2- EBC after locoregional tx and neoadjuvant CT3 1. Johnston. JCO. 2020;38:3987. 2. Rastogi. SABCS 2020. Abstr GS1-01. 3. Loibl. SABCS 2020. Abstr GS1-02. 313 Why Different Outcomes Across These Trials (or Are There)?  Different definitions of high risk? ‒  Was there more luminal B (high proliferation) in monarchE than in PALLAS or PENELOPE-B? Differences in therapy adherence? ‒ May explain PALLAS results but adherence much higher in PENELOPE-B (though shorter duration of therapy with only 1 yr)  Is abemaciclib a more effective CDK inhibitor? ‒ Possible, although not supported by metastatic first-line trials that have remarkably similar hazard ratios (continuous dosing, more potent inhibition of CDK4, monotherapy activity)  Durations of CDK inhibitor therapy? ‒ Possible PENELOPE-B would have been positive if palbociclib had been given for longer ‒ Await NATALEE results with 3 yr of ribociclib O’Regan. SABCS 2020. Abstr GS1-03. Why Should Neoadjuvant Endocrine Therapy Be Given?  Down-stage tumor prior to surgery ‒ NET and chemotherapy have similar rates of clinical response, radiographic response, and rates of BCS, but lower rates of toxicity ‒ AI superior to tamoxifen in terms of clinical response, radiographic response, and rates of BCS ‒ Longer duration of NEC allows for higher rates of BCS and pCR  Identify HR+ tumors that may not require chemotherapy  Gain insights into the biology of the tumors ‒ Biomarkers response/resistance to treatment ‒ Predict phase III trial results Barchiesi. Int J Mol Sci. 2020;21:3528. 314 No Significant Improvement in ORR/BCS With Neoadjuvant Chemotherapy vs Endocrine Therapy Objective Response Breast Cancer Survival 3 mo Objective response by clinical palpation A: 63% B: 64% A: 24% B: 33% P = .058 (statistically significant) GEICAM 2006-03 A: CT (EC -> doxetaxel) B: Exe (+ goserelin if premenopausal) -- Response rate by MRI A: 66% B: 48% A: 47% B: 56% NEO CENT A: CT (n = 22) B: Letrozole (n = 22) 18-23 wk Recruitment feasibility and tissue collection A: 54% B: 59% -- A: CT (n = 53) B: Letrozole/ palbociclib (n = 53) 20 wk Residual cancer burden index A: 76% B: 75% A: 69% B: 69% Trial Treatment Arm Duration Primary Endpoint Semiglazov et al. A: CT (n = 118) (dox + paclitaxel) B: ET (total n = 121) Ana (n = 60) + Exe (n = 61) UNICANCER-NEO Pal Reinert. Curr Treat Options Oncol. 2018;19:23. Neoadjuvant ET vs CT: Systematic Review/ Meta-analysis of 20 RCTs Through 2015 (N = 3490)  AIs showed significantly higher rates of clinical response, radiologic response, and BCS vs tamoxifen; radiologic response rate higher with CT + ET vs ET Clinical Response Source Favors Favors Endocrine Chemotherapy OR (95% CI) Pathologic CR Source Alba 2012 2.11 (0.92-4.82) Alba 2012 Palmieri 2014 0.34 (0.6-1.98) Palmieri 2014 Semiglazov 2007 0.93 (0.55-1.57) Semiglazov 2007 1.84 (0.53-6.47) Total 1.08 (0.50-2.35) Total 1.99 (0.62-6.39) Heterogeneity: x22 = 4.47 (P = .11), I2 = 55%. Test for overall effect: z = 0.19 (P = .85). 0.01 Radiologic Response Source 0.1 1.0 10 OR (95% CI) 100 Favors Chemotherapy 3.13 (0.12-78.77) Not estimated Heterogeneity: x22 = 0.09 (P = .76), I2 = 0%. Test for overall effect: z = 1.16 (P = .25). 0.01 0.1 1.0 10 OR (95% CI) 100 0.01 0.1 1.0 10 OR (95% CI) 100 Breast Conservation Response OR (95% CI) Alba 2012 2.11 (0.92-4.82) Source Palmieri 2014 0.83 (0.25-2.74) Alba 2012 0.68 (0.30-1.54) Semiglazov 2007 1.28 (0.77-2.14) Semiglazov 2007 0.63 (0.36-1.11) Total 1.38 (0.92-2.07) Total 0.65 (0.41-1.03) Heterogeneity: x22 = 1.77 (P = .41), I2 = 0% Test for overall effect: z = 1.54 (P = .12) Favors Endocrine OR (95% CI) 0.01 0.1 1.0 10 OR (95% CI) 100 OR (95% CI) Heterogeneity: x22 = 0.03 (P = .87), I2 = 0% Test for overall effect: z = 1.83 (P = .07) Spring. JAMA Oncol. 2016;2:1477. 315 Summary of Endocrine Therapy Alone  Neoadjuvant endocrine therapy yields similar ORR response to chemotherapy ‒ AI > tamoxifen in terms of ORR and BCS  Endpoints of trials in ER+ breast cancer challenging ‒ Achievement of pCR rare (<5%) ‒ Use of Ki67 and PEPI may correlate with long-term outcome and may allow triage of patients with no drop in Ki67 to chemotherapy ‒ ALTERNATE: fulvestrant = AI = AI + fulvestrant in terms of Ki67 response, PEPI; long-term outcomes are awaited neoMONARCH: Neoadjuvant Abemaciclib, Anastrozole, and Abemaciclib + Anastrozole in HR+/HER2- BC  RNAseq analysis of biopsy samples from multicenter, randomized, open-label phase II trial 2-wk lead-in Postmenopausal women with stage I-IIIB HR+/HER2- BC B I O P S Y 14 wk Abemaciclib + ANZ (n = 11)* B I O P S Y Abemaciclib (n = 21) ANZ (n = 17) Abemaciclib + ANZ (n = 10) Abemaciclib + ANZ (n = 16) Abemaciclib + ANZ (n = 17) B I O P S Y n=8 n=9 n=9 n = number of biopsy samples.  Primary endpoint: percent change in Ki67 from baseline to 2 wk of treatment  Secondary endpoints: pCR, OR, radiologic response Hurvitz. Clin Cancer Res. 2020;26:566. 316 neoMONARCH: Antiproliferative Effects of Abemaciclib, Anastrozole, and Combination Tx on HR+/HER2- BC Response per CCCA (Ki67 ≤2.7%) at Wk 2 -20 -40 -60 -80 -100 Outcome, % Abema + ANZ Abema ANZ Geometric mean change in Ki67 at 2 wk -93 -91 -63 40 CCCA 68 58 14 20  ORR: 46% (5% CR, 42% PR) 100 0 Patients With Tumors Exhibiting CCCA (%) Geometric Mean Change in Ki67 (%) Change in Ki67 From BL to 2 Wk P <.001 P <.001 80 60 0 P <.001 P <.001 Abemaciclib + ANZ (n = 59) Abemaciclib (n = 52) ANZ (n = 56) Abemaciclib + ANZ (n = 40) Abemaciclib (n = 30) ANZ (n = 8)  pCR: 4% Hurvitz. Clin Cancer Res. 2020;26:566. PALLET: Palbociclib + Neoadjuvant Letrozole in ER+/HER2- EBC  Randomized phase II trial (parallel protocols in North America and UK) Stratified by country Postmenopausal women with ER+/HER2-invasive EBC, tumors ≥2 cm by ultrasound, no metastatic disease (N = 307) Group A Letrozole* for 14 wk Group B Letrozole for 2 wk → Letrozole + palbociclib† to wk 14 3:2:2:2 Group C Palbociclib for 2 wk → Letrozole + palbociclib to wk 14 Group D Letrozole + palbociclib for 14 wk *Letrozole 2.5mb/day PO. †Palbociclib 125 mg/day PO (3 wk on, 1 wk off). Dose reduction to 100 mg and 75 mg available.  Coprimary endpoints (for group A vs B + C + D): change in Ki67 (protein between baseline and 14 wk and clinical response (ordinal and Johnston. JCO. 2019;37:178. ultrasound) after 14 wk  317 S U R G E R y Follow-up 30 days after last treatment and 12 mo after randomization Core cut biopsies taken at:  Baseline (post randomization)  2 wk (prior to start of second drug in groups B and C)  14 wk or at discontinuation treatment (within 48 hr of last dose) Secondary endpoint: pCR PALLET: Clinical Response (Coprimary Endpoint) Response Categories Patients in Response Category (%) 100 80 5.4 3.2 1.6 4.8 3.2 45.2 47.6 41 38.7 42.5  No significant difference in ORR with letrozole (Group A) vs letrozole + palbociclib (Groups B + C + D) 60 40 54.8 47.3 0 54.1 47.6 20 2.2 LET alone 14 wk (Group A) 1.6 3.3 LET 2 wk then LET+PAL 12 wk (Group B) PAL 2 wk then PAL+LET 12 wk (Group C) ■ PD 52.2 ‒ ORR: 49.5% vs 54.4%; P = .20 per Mann-Whitney test (ordinal) ■ SD 1.6 ■ PR PAL+LET 14 wk (Group D) 2.2 PAL+LET regimens (Groups B+C+D) ■ CR Johnston. JCO. 2019;37:178. PALLET Summary  Compared to letrozole, palbociclib + letrozole: ‒ Enhanced Ki67 suppression (P <.001) ‒ Increased CCCA rate (58.5% to 90.4%; P <.001) ‒ Did not improve ORR (49.5% to 54.4%; P = .2) ‒ pCR rate: 1% Johnston. JCO. 2019;37:178. 318 FELINE: Neoadjuvant Ribociclib + Letrozole vs Placebo + Letrozole in ER+/HER2- Breast Cancer  Randomized, open-label phase II trial (patients accrued Feb 2016 to Aug 2018)  6 28-day cycles Postmenopausal women with HER2- breast cancer, stage II/III, primary >2 cm; ER+ >66% (N = 120) Ribociclib 400 mg/day PO, continuously Letrozole 2.5 mg PO QD (n = 41)  Surgery between D8-21 of cycle 6 Ribociclib 600 mg/day PO, 3 wk on/off Letrozole 2.5 mg PO QD (n = 41)  Clinical response measurement: ultrasound, MRI Letrozole 2.5 mg PO QD + Placebo (n = 38)  Treatment continued until day before surgery  Tissue samples: baseline, Day 14 of cycle 1, and at surgery  If Ki-67 > 10% at Day 14 of cycle 1, patient removed from trial  Primary endpoint: Proportion reaching PEPI score 0 at surgery (ie, removing need for adjuvant chemo) − PEPI 0: tumor ≤5 cm, node negative, Ki-67 ≤2.7%, Allred ER score 3-8  Secondary endpoints: complete cell-cycle arrest, responses (RECIST), safety Khan. ASCO 2020. Abstr 505. FELINE: Ki67 Change Between Baseline and Day 14 of Cycle 1 Group A: Change in Ki67 (Baseline to D14C1) 80 Group A Placebo + Letrozole Groups B+C P Value Group B Ribociclib Intermittent + Letrozole Group C Ribociclib Continuous + Letrozole Baseline Ki67, median % 15.8 21.4 .9129 16.5 24.7 Day 14 Ki67 > 10%, % (n/N) 17.24 (5/29) 4.05 (3/74) .025 2.86 (1/35) 5.13 (2/39) Mean change Ki67% (baseline to D14C1) -15.7 -23.3 .047 -20.6 -25.7 CCCA at Day 14, % (n/N) 51.7 (15/29) 91.9 (68/74) <.0001 97.1 (34/35) 87.2 (34/39) 40 20 0 BL Day 14 Groups B+C: Change in Ki67 (Baseline to D14C1) 80 60 Ki67 (%) Parameter Ki67 (%) 60 40 20 0 BL Khan. ASCO 2020. Abstr 505. 319 Day 14 FELINE: Summary  Adding ribociclib to letrozole did not  Increase number of patients with PEPI = 0  Improve ORR  Adding ribociclib to letrozole did  Improve complete cell cycle arrest at C1D14  But this was not maintained at surgery (acquired resistance?) NeoPal: Neoadjuvant Letrozole + Palbociclib vs CT  Randomized, parallel, non-comparative phase II trial Patients with ER+/HER2- BC; Prosigna-defined luminal B or luminal A/N+; stage II-III; not candidate for breast conserving surgery (N = 106) Letrozole 25 mg QD + Palbociclib 125 mg QD 3/4 wk x 19 wk (n = 53) FEC100* x 3 cycles then Docetaxel 100 mg/m2 x three 21-day cycles (n = 53) S U R G E R y Final Analysis Outcome, n (%) LET + PAL (n = 52) CT (n = 51) RCB 0-I 4 (7.7) 8 (15.7) RCB II-III 48 (92.3) 43 (84.3) 2 (3.8) 3 (5.9) pCR *FEC100: 5-FU 500 mg/m2, epirubicin 100 mg/m2, cyclophosphamide 500 mg/m2.  Primary endpoint: rate of RCB 0-I  Secondary endpoints: clinical response, proliferation-based markers, safety  Cottu. Ann Oncol. 2018;29:2334. 320 Sample size: null hypothesis (p0) was RCB 0-I in 20% of cases; alternative hypothesis was 40%, type I error of 0.045, type II error of 0.042 (power: 95.8%); required sample size n = 60/arm NeoPal Conclusion  Neoadjuvant LET + PAL or CT was associated with poor pathologic response for Prosigna-defined high-risk luminal breast cancer – RCB 0-1 rate: 7.7% with LET + PAL; 15.7% with CT – pCR rate: 3.8% with LET + PAL; 5.9%% with CT  Neoadjuvant LET + PAL or CT led to similar rates of clinical or radiologic response and BCS  PEPI = 0 was numerically higher with LET + PAL (17.6% vs 8.0% with CT) Cottu. Ann Oncol. 2018;29:2334. CORALLEEN: Neoadjuvant Letrozole + Ribociclib vs CT  Multicenter, randomized, open-label phase II trial Stratified by tumor size (T1/2 vs T3), nodal involvement Postmenopausal women with HR+/HER2- BC; stage I-IIIA; tumor size ≥2 cm; PAM50 luminal B (Prosigna) (N = 106) Letrozole 2.5 mg/day + Ribociclib 600 mg/day for 3/4 wk (n = 52) AC* Q3W x 4 Paclitaxel 80 mg/m2 wkly x 12 (n = 54)   Surgery performed within 7 days of last dose of ribociclib or within 2 wk of last dose of CT ET in the ribociclib + letrozole group continued until day of surgery *Doxorubicin 60 mg/m2 + cyclophosphamide 600 mg/m2. PAM50 + RNA/DNA seq and plasma samples collected at baseline, Day 15, and post surgery.  At baseline, ~40% of patients had node-positive disease, median Ki67 expression was 30-35, median ROR score was 70-77, and ≥85% had ROR high-risk disease Prat. Lancet Oncol. 2020;21:33. 321 CORALEEN: Primary Endpoint ROR Ribociclib + Letrozole (n = 49) Chemotherapy (n = 52) n (%) 95% CI n (%) 95% CI Low 23 (46.9) 32.5-61.7 24 (46.1) 32.9-61.5 Intermediate 15 (30.6) 18.2-45.4 13 (30.8) 19.1-45.9 High 11 (22.5) 11.8-36.7 11 (21.2) 11.2-35.2 NA NA 1 (1.9) NA Missing Prat. Lancet Oncol. 2020;21:33. Summary: Neoadjuvant CDK4/6 Inhibitors  Addition of CDK4/6 inhibitors to ET shows biologic activity (potent reduction in Ki67 at 2 wk)  Addition of CDK4/6 inhibitors to ET does not appear to improve ORR or pathologic response vs ET alone or CT, with low rates of pCR or RCB 0-1  Cross-trial comparisons suggest similar level of benefit across the 3 CDK4/6 inhibitors; however, there was no head-to-head comparison  The optimum endpoint is unknown; it has not been firmly established that increasing the rate of pCR and RCB 0-1 improves long-term outcomes for luminal cancers  Without long-term outcomes, neoadjuvant CDK4/6 inhibitors remain investigational 322 Summary  Use of genomic assays in the preoperative setting may help us best select patients who may benefit from preoperative endocrine therapy vs chemotherapy  Assessing endpoint to assess response to endocrine therapy + CDK4/6 inhibition not clearly established 323 Metastatic breast cancer– standards and perspectives Simona Borštnar Division of Medical Oncology Institute of oncology Ljubljana 2nd Summer School in medical oncology September 2021 Targeted treatment in MBC Anti-HER2 therapy (trastuzumab, pertuzumab, lapatinib, T-DM1, T-Dx, tucatinib HER2+ gBRCA1/2 mut TNBC HR+,HER2PIK3Ca mut 324 HR+ HER2- MBC Peri/premenopausal VISCERAL CRISIS: leptomeningeal carcinomatosis; hepatic metastases with endangered liver function: increasing bilirubin levels with biliary obstruction excluded, elevated liver transaminases with diffuse hepatic infiltration; pulmonary lymphangitic carcinomatosis; large lung metastases with respiratory insufficency present YES Visceral crisis1 NO ChT Postmenopausal Until best response Ovariectomy or MOS Low disease burden, long disease free interval YES NO AI or fulvestrant or tamoxifen AI+ CDK4/6i , or fulvestrant + CDK4/6i if <12 months from adjuvant AI PIK3CA mut. NO BRCA1/2 mut. YES NO YES ful + alpelisib PARPi Everolimus + ET(e.g.. eksemestan, tamoksifen), not used Ai (steroidal or nonsteroidal) fulvestrant, megestrol acetat CHT (monotherapy , not used before) HER2 +MBC No prior treatment with anti-HER2 therapy Prior (neo) adjuvant treatment with anti-HER2 therapy Taxanes+ trastuzumab + pertuzumab Taxanse + trastuzumab + pertuzumab (if the free disease interval is > 12 months) or T-DM1 (if the free disease interval is< 12 months or ChT1 + trastuzumab Patients unsuitable for CT or long free interval and minimal disease extent, HR+ ET+ anti-HER2 therapy (trastuzumab ali lapatinib or trastuzumab+pertuzumab) Without progression Maintenance anti-HER2 therapy Disease progression The optimal duration of maintenance anti-HER2 therapy is unknown; possible to stop after several years of complete remission of the disease. T-DX Tukatinib + trast.+ capecitabin 3 capecitabin+ lapatinib T-DM1 CHT3+or HT4 +trastuzumab 325 1 noncardiotoxic CT (taxanes, vinorelbine, paclitaxel / carboplatin) 2 non-cardiotoxic CT not previously used 3 preferred in CNS metastases 4 in HR + HER2 + BC trastuzumab + lapatinib TRIPLE NEGATIVE ABC Tumor testing for PD-L1, Testing for g BRCA1/2 mutation PD-L1 neg PD-L1 pos Atezolizumab+ nabpaklitaksel BRCA1/2 neg BRCA1/2 pos Rapidly progressing disease, visceral crisis YES NO monotherapy without prior treatment with anthracyclines and taxanes PARP inhibitors polichemotherapy Previously treated with anthracyclines and taxanes 1 with anthracyclines can be treated up to the maximum total dose 2 taxanes can be reused if the free disease interval after (neo) adjuvant treatment is> 12 months Anthracyclines or taxanes carbo/cisplatine capecitabine vinorelbine eribulin Other unused ChTs, selection according to previous efficacy, side effects, performance status Conclusion  Approximately 20% to 30% of woman initially diagnosed with early stage disease will develop metastatic breast cancer.  The goals of treatment are: maximizing the quality of life, prevention and palliation of symptoms, and prolongation of survival. Treatment is lifelong, characterized by remissions and relapses.  When choosing a treatment, it is necessary to know the tumor characteristics and identify molecular targets.  Targeted drugs such are anti-HER2 therapies in HER2 positive subtype, CDK4/6, mTOR and PIK3CA inhibitors in HR+/HER2- subtype and immune check point inhibitors in TNBC have significantly improved disease control.  New promising drugs are under way, however we need clinical trials and continued basic and translational research to make new breakthroughs. 326 The diffuse large B cell lymphoma – recommendations and current knowledge Barbara Jezeršek Novaković Onkološki inštitut Ljubljana Conclusion LBCL are a heterogeneous group of diseases with different clinical and biological features – for an adequate treatment, histopathological evaluation of an extirpated lymph node is a prerequisite. 327 Conclusion With current treatment, only 50 to 60% of patients will be cured. In relapsed disease, salvage chemotherapy results in long term disease free survival in a small minority of patients. Additional curative option is offered through high dose therapy and autologous SCT. Multiply relapsed lymphomas can be treated with CAR T cell therapy and allogeneic SCT, other treatments are predominately palliative. And results… Oncol Lett. 2018 Mar; 15(3): 3602–3609. Published online 2018 Jan 11. doi: 10.3892/ol.2018.7774 PMCID: PMC5796369 Diffuse large B-cell lymphoma: 10 years' real-world clinical experience with rituximab plus cyclophosphamide, doxorubicin, vincristine and prednisolone Matej Horvat,1 Vesna Zadnik,2 Tanja Južnič Šetina,1 Lučka Boltežar,1 Jana Pahole Goličnik,1 Srdjan Novaković,3 and Barbara Jezeršek Novaković1 328 And results… Table I. Patient clinical and demographic characteristics at the start of treatment (n=624). Sex Male 297 (48%) Female 327 (52%) Median age 67.0 (19-89) <60 years, 208 (33%) ECOG performance status 0: 299 (48%) 1: 183 (29%) 2: 90 (14%) 3: 33 (5%) 4: 19 (3%) Disease stage I: 73 (12%) II: 178 (29%) III: 111 (18%) IV: 245 (39%) Elevated LDH level 311 (50%) Extranodal involvement 439 (70%) Treatment regimen Rituximab + CHOP 575 (92%) Rituximab + other chemotherapy 32 (5%) Chemotherapy alone 17 (3%) IPI score 0: 63 (10%) 1: 122 (20%) 2: 143 (23%) 3: 141 (23%) 4: 108 (17%) 5: 44 (7%) And results… 329 And results… Survival rate (%) 1-year 2-year 3-year 5-year 10-year Median survival (months) Age, <60 years 88 85 84 83 79 NR Age. ≥60 years 84 80 79 78 70 NR 93 90 89 87 77 NR 83 79 79 79 79 NR Age, <60 years 93 85 81 81 76 NR Age, ≥60 years 82 70 64 56 41 80.1 98 95 91 91 87 NR 87 76 71 71 67 NR Outcome Progression-free survival Age, <60 years, IPI 0 or 1 Age, <60, IPI ≥2 Overall survival Age, <60 years, IPI 0 or 1 Age, <60, IPI ≥2 And results… 330 Marginal zone lymphoma (MZL) Summer school 2021 Oncology institute of Ljubljana Milica Miljković, medical oncologist Introduction Indolent (slow growing) Non – Hodgkin B cell lymphomas 5-15% of Non-Hodgkin lymphomas Average age at diagnosis is 60 years Slightly more common in women 331 WHO classification Clonal B-cell lymphocytosis of marginal zone origin (CBL-MZ) Nodal marginal zone lymphoma (NMZL) Splenic marginal zone lymphoma (SMZL) Extranodal marginal zone lymphoma (EMZL) or Mucosa –Associated Lymphoid Tissue (MALT) : The stomach is the most common site, followed by ocular adnexa, lung and salivary glands. Treatment of EMZL, NMZL, SMZL Treat just symptomatic systemic disease It means: -B simptomps: fewer, night sweats and weight loss -Bulky disese -Symptomatic splenomegaly and/ or any progressive cytopaenias - Hb < 100 g/L - Platelets < 80x10/L - Neutrophils < 1 x10/L 332 Treatment of EMZL, NMZL, SMZL Stage I and II: surgery +/- RT (25,2GY) Stage III and IV and with B simptoms: 6-8 x R-LP (rituximab,chlorambucil, prednisolone) 4-6 x R-Bendamustine 6-8 x R-Lenalidomide 4 x Rituksimab + RT for old and unfit patients 6-8 x R-CHOP for aggressive disease (rituximab,ciklofosfamid,doxorubicin,vincristine,prednisolone) Treatment algorithm for localised gastric MZL 333 Treatment algorithm for advanced gastric MZL 334 Sekcija internistične onkologije pri SZD, Onkološki inštitut Ljubljana in Katedra za onkologijo 2nd Summer School in medical oncology – Precision oncology – Are we there yet 7-10 September 2021 Institute of Oncology Ljubljana, Zaloška 2, Ljubljana Standards and perspectives in the systemic treatment of Lymphomas – Follicular lymphoma dr. Tanja Južnič Šetina, dr.med Institute of Oncology, Ljubljana Follicular lymphoma • Follicular lymphoma (FL) is the second most common subtype of non-Hodgkin lymphoma • It accounts for 20% of all NHL cases and around 70 % of indolent lymphomas • The clinical presentation is usually characterized by asymptomatic peripheral lymphadenopathy in cervical, axillary, inguinal and femoral regions. Most patients are diagnosed with advanced stage disease. Bone marrow involvement is present in more than >80%. • The course of FL is quite variable. The disease is usually characterized by an indolent clinical course and response to initial therapy, but relapses are common. • The prognosis of FL is good. The median survival of FL is over 15 years but considering it`s indolent nature, survival can often be measured in decades. • Histologic transformation of FL to a DLBCL has been reported in up to 70 percent of patients over time, and is associated often with a poor prognosis. 335 PRIMA – PI • B2M increased • Bone marrow involvement • • • FLIPI - outcome according to risk group 5 year OS % 10-year OS % Low Risk group 0 to 1 score 91 71 Intermediate 2 78 51 High >3 52 36 score 1. 2. 2- yr OS % 2-yr PFS % mPFS (months) 98 84 84 2 94 72 70 >3 87 65 42 OS by FLIPI risk groups¹ OS by FLIPI risk groups for R-containing regimens² FLIPI score applied to patients in the rituximab era (68% received R, n=2192) 0 to 1 low risk: B2M normal and bone marrow not involved intermediate risk: B2M normal and bone marrow involved high risk: B2M elevated Solal-Celigny, P, Roy, P, Colombat, P, et al. Follicular lymphoma international prognostic index. Blood 2004; 104:1258 Nooka AK, Nabhan C, Zhou X, et al.. Ann Oncol 2013; 24:441. 336 Initial treatment of stage I to IV follicular lymphoma  treatment of FL depends on the stage of disease at presentation  introduction of anti-CD20 monoclonal antibody rituksimab into first line FL treatment or relapsed disease significantly improved the outcome of FL patients Stage I/II • 10-20% of FL patients present with limited stage I/II disease • radiotherapy (24-30 Gy) is generally the treatment of choice for limited stage FL, and results in 10-year OS rates of 60% to 80%; • 2 x 2 Gy schedule is less durable but might be used in special situations to minimize side-effects (e.g. lacrimal gland, parotid glands) • In selected cases (e.g. limited life expectancy, large abdominal fields), observation or rituximab monotherapy may be considered Stage III/IV • Initial observation ("watch and wait" ) is the standard approach in asymptomatic, stable patients with stage II (high tumor burden), III, or IV • Immunotherapy-based treatment (rituximab, obinutuzumab and chemotherapy) • Radioimmunotherapy • lenalidomide-rituximab • rituximab monotherapy or R in combination with chlorambucil Indications for treatment • local symptoms due to progressive or bulky nodal disease • vital organ compression • presence of systemic B symptoms ( fevers >38°C, weight loss, night sweats) • presence of symptomatic extranodal disease, such as effusions • cytopenias due to extensive bone marrow infiltration, autoimmune hemolytic anemia or thrombocytopenia, or hypersplenism • rapid lymphoma progression 337 First-line therapy • Preferred regimens • R – bendamustin, R-CHOP or R-CVP • Obinutuzumab / bendamustin, CHOP or CVP • Rituximab+ lenalidomide • Less preferred • Rituximab – monotherapy (4 weekly doses) • Rituximab+chlorambucil • Radioimmunotherapy • Rituximab maintenance (every 8 weeks for 2 years) - improves PFS and OS • Obinutuzumab maintenance (every 8 weeks, 12 doses) Randomized trials in first-line FL treatment (R-chemotherapy) Study Treatment, n Median FU, months ORR, % PFS % Median TTP/ TTF/ EFS, mo OS, % Marcus et al. 2006 CVP, 159 R-CVP, 162 53 57 81 15 34 p<0.0001 77(4-y) 83 (4-y) p = 0.029 Hiddemann et al. 2005 CHOP-IFN, 205 R-CHOP-IFN, 223 18 90 96 29 NR p<0.001 90(2-y) 95 (2-y) p = 0.016 Herold et al. 2006 MCP-IFN, 96 R-MCP-IFN, 105 47 75 92 71 26 NR p<0.0001 74 (4-y) 87 (4-y) p = 0.009 Salles et al.2008 CHVP-IFN, 183 73 37 84 53 33 p<0.0004 67 79(5-y) p = 0.07 84 (5-y) 69 70 (10-y) 31 p<0.0001,HR 0.58 66 (10-y) Schulz H, 2007 metaanalysis 60 R-CHVP-IFN, 175 PFS: R-ChT vs ChT ( HR 0,62, CI 0,55-0,71, p<0,001) OS: R-ChT vs ChT, 1480 (1.line+relapses) (HR 0,63; 95 % CI 0,51-0,79, p<0,001) 92,7 R-Bendamustine, 279 Rummel et al. 2009 Morschhauser et al. 2018 R- CHOP, 275 R-CHOP/BR + R maint., 517 R-lenalidomide + R maint., 513 45 38 84 38 89 PFS 78% (3y) 77% (3y) 338 94 (3 y) 94 (3 y)  With the introduction of R, survival of FL has improved.  The improvement was established in at least four prospective firstline trials and a systematic metaanalysis. Maintenance treatment with rituximab (PRIMA study) PFS (PRIMA) 1. Long-Term Results of the PRIMA Study (9y FU) Study FU, n Treatment Median PFS OS PRIMA, 2011 First-line FL 9y of FU R-ChT+observation vs R-ChT+mainten R 4.1y >80 (10-y) 10.5y p<0,001 >80 (10-y) p= NS 1018 pts CONCLUSION (PRIMA) • rituximab maintenance after induction immunochemotherapy provides a significant PFS benefit over observation • no OS benefit OS (PRIMA) 2. Meta-analysis (Vidal et al, 2017); seven trials including PRIMA, 2315 pts with FL evaluating rituximab maintenance on OS CONCLUSION: Maintenance rituximab improved PFS (HR 0.57; 95% CI 0.510.64) and OS (HR 0.79; 95% CI 0.66-0.96) 1. Bachy E, et al. J Clin Oncol. 2019;37(31):2815-2824. Sustained Progression-Free Survival Benefit of Rituximab Maintenance in Patients With Follicular Lymphoma: Long-Term Results of the PRIMA Study. 2. Vidal L, Gafter-Gvili A, Salles G, et al. Eur J Cancer. 2017;76:216 GALLIUM Study (randomised phase III study in previously untreated FL patients) Induction Grade 1–3a ECOG PS 0–2 (N=1202) R R-chemo (601 pts) R 375mg/m2 IV on D1 of C1–8 (q3w) or C1–6 (q4w) plus CHOP,CVP or bendamustine Primary endpoint • PFS CR or PR† at EOI visit Previously untreated FL Stage III/IV or stage II bulky (≥7cm) requiring treatment Maintenance G-chemo (601 pts) G 1000mg IV on D1, D8, D15 of C1 and D1 of C2–8 (q3w) or C2–6 (q4w) plus chop,CVP or bendamustine Follow-up G G 1000mg IV q2m for 2 years or until PD Median follow-up: 34 months R R 375mg/m2 IV q2m for 2 years or until PD Marcus, et al NEJM 2017; 377(14):1331-1344 339 GALLIUM study results : PFS and OS in patients with FL (first-line) R-ChT, n=601 3-y PFS,% (95% CI) HR (95% CI), =p=0.001 73,3 (68.8-77.2) 3-y OS HR 0.75, P=0,21 G-ChT, n=601 80,0 (75.9-83.6) 92 94 Conclusion:  Obinutuzumab/chemo + maintenance is superior to similar R-based regimens with improvement of PFS  no difference in OS Marcus, et al NEJM 2017; 377(14):1331-1344 RELEVANCE: Phase III Trial Design (previously untreated FL patients) Treatment period 1 (~6 months) Treatment period 2 (~1 year) Treatment period 3 (~1 year) R2 Rituximab n = 513 Previously untreated advanced FL requiring treatment per GELF (N = 1,030) R2 1:1 R-chemo n = 517 Rituximab (R-CHOP, R-B, R-CVP) R2 = lenalidomide/rituximab; R = rituximab; B = bendamustine; CVP = cyclophosphamide/ vincristine/prednisone Total treatment duration: 120 weeks Primary endpoints: CR/CRu at 120 weeks and PFS Morschhauser F, Fowler NH, et al. N Engl J Med. 2018;379(10):934 340 RELEVANCE: Interim PFS (first-line FL patients) Interim PFS R2 (n = 513) 3-year PFS R-chemo R-chemo (n = 517) 77% 78% HR 1.10 p-value 0.48 Conclusions • similar rates of CR (48% in R2 vs 53 % in R chemo) • similar 3-y PFS (77% vs 78 %, HR 1.10, 95% CI 0.85-1.43) • similar OS (94 vs 94 %, HR 1.16, 95% CI 0.72-1.86) • more rash, diarrhea and tumor flare, less neutropenia in the R2 R2 • At median follow-up of 37.9 mo, interim PFS was similar in both arms • 3-y OS = 94% (R2) vs 94% (R-chemo), HR 1.16 Morschhauser F, Fowler NH, et al. N Engl J Med. 2018;379(10):934. Treatment of R/R FL • 20 to 30 % of patients with FL relapse after first- line therapy • rule out histologic transformation • patients who progress within 24 months have significantly worse OS than those who do not • asymptomatic recurrent FL do not necessarily require immediate treatment 341 Treatment regimens for R/R FL • Preferred regimens • • • • Bendamustin + rituximab or obinutuzumab CHOP or R-CVP + obinutuzumab or rituximab Lenalidomide + rituximab or obinutuzumab PI3K inhibitors (after 2 prior therapies) • Less preferred (eldery, frail pts) • • • • Rituximab alone (4 weekly doses) Chlorambucil +/- R Cyclophosphamide +/- R Radioimmunotherapy • Rituximab maintenance (every 3 months for 2 years) • Obinutuzumab maintenance for R-refractory (every 8 weeks, 12 doses) • Other • High dose therapy with ASCT, allogeneic transplant for selected patients • Novel treatments (CAR-T-cell therapy, tazemetostat – EZH2 mut posit after 2 prior therapies or EZH2 WT in pts with no other alternative), mosunetuzumab R, rituximab Randomised phase III study for R/R FL patients + 342 Obinutuzumab maintenan. (1000 mg every 2 months) for up to 2 years OS (HR, 0.67; 95% CI, 0.47 to 0.96; P = .027) median follow-up 31,8 months Conclusion: the addition of obinutuzumab improves PFS and OS. Toxicity was similar in both treatments. Cheson BD. J Clin Oncol. 2018;36(22):2259. AUGMENT: R2 versus Rituximab/Placebo for R/R FL or Marginal Zone Lymphoma - randomised phase III study 343 Conclusions: • improved PFS with R2 vs R alone (39 mo vs 14 mo) • 2-year OS was 95% for R2 and 86% for R/placebo • similar efficacy to chemoimmunotherapy Idelalisib • oral PI3K inhibitor that has shown therapeutic activity in initial studies of patients with multiple relapsed FL • approved by EMA and FDA as a single agent for the treatment of patients with relapsed FL who have received at least two prior systemic therapies • approval based on a phase 2 study of idelalisib (n=72) in patients with FL who have received at least 2 prior lines of therapy1 • ORR was 57 % with a median duration of response of 12.5 months • The starting dose is 150 mg twice daily • toxicity: • diarrhea – 14% • cytopenias – 28% • infections – 21% pneumonitis - 4% hepatotoxicity – 18% hypertriglyceridemia, hyperglycemia • anti-infectious prophylaxis 1.Gopal AK, et al. N Engl J Med. 2014 344 Avtologous, allogeneic transplantation • an appropriate consolidative therapy for patients in second or third remission • may have the greatest benefit in patients with early treatment failure, refractory FL and those with histologic transformation to a more aggressive histology • there is a plateau in the survival curves between 10 and 15 years after autologous HCT, suggesting that this procedure may be curative for one-quarter to one-third of transplanted patients1,2 • allogeneic transplantation can be considered in highly selected patients (lower rates of relapse vs autoHCT, high TRM up to 20%) 1. Metzner B, et al. Ann Oncol. 2013 2. Casulo C, et al. Biol Blood Marrow Transplant. 2018 Conclusions • FL is a chronic, incurable disease with a long natural history • a small subgroup of patients presenting with limited stage disease may be cured with radiation • observation is the first approach in asymptomatic patients • for those who require therapy : multiple treatment regimens, anti-CD20 +/- chemotherapy and chemotherapy-free alternatives, novel agents,… o antiCD20 +/- chemotherapy o lenalidomide +/- rituximab o PI3K inhibitors o autologous and allogeneic stem cell transplantation o novel agents (tazemetostat - EZH2 inhibitor, mosunetuzumab - bispecific antibody ,…) o CAR-T cell therapy - may dramatically alter the prognosis for heavily pretreated patients (ZUMA-5 study with high ORR more than 90%, median PFS at 2 years 78%, CR rate 80 %,..) • disease can still be fatal in patients with histologic transformation 345 MANTLE CELL LYMPHOMA Monika Jagodic, MD, PhDmmd SUMMARY: Mantle cell lymphoma (MCL) Histology: the image is of mantle zone cells surrounding normal germinal centre follicles Biology: the translocation t (11;14) (q13; q32) leading to cyclin-D1 overexpression is typical Prognosis: Clinical and biological prognosticators (combined mantle cell lymphoma International Prognostic Index, MIPI-c) should be used routinely to estimate clinical behaviour) including age, ECOG, LDH, WBC count, Ki-67 index. Other prognosticators: SOX-11 expression, TP 53 mutations Initial treatment approach: a short course of watch-wait under close observation is recommended for suspected indolent cases with low tumour burden and asymptomatic disease at diagnosis. When starting of treatment is required (symptoms, high tumour burden) initial treatment is conventional chemoterapy (ChT) with rituximab (Rx) +/-consolidation RT. In younger patients are used aggresive regimens by autologous stem-cell transplantation (SCT) as 1st line. In older patients are used conventional ChT combinations and less intense immunoChT or low- toxicity single agent targeted therapy in frail patients. Maintenance with rituximab significantly improves progression-free survival and overall survival. Relapse disease treatment: salvage therapy: in early relapses (<12-24 months) a non-cross resistant scheme after R-CHOP or vice versa is recommended, with Rx maintenance. Molecular targeted substances (ibrutinib, lenalidomide) should be considered in refractory disease. Additional options: temsirolimus, bortezomid. AlloSCT transplantation should be considered in younger patients as potentially curative treatment in early relapse and refractory disease. 346 T-CELL LYMPHOMA Jana Pahole MD Institute of Oncology Ljubljana 10/09/2021 T-CELL LYMPHOMA – short facts • • • • 7-10% of all NHL Most are clinically aggressive Heterogenous in their presentation, features and prognosis Lack of efficient treatment • Most can be healed not cured • Outcomes are poor • • • • Low response rates 5-year OS ~32% 5-year failure-free survival ~20% Most patients relapse within 2-3 years 347 T-CELL LYMPHOMA SUBTYPES LEUKEMIC • T-cell prolymphocite leukemia • Adult T-cell leukemia/lymphoma • T-cell large granular lymphocytic leukemia • NK cell leukemia NODAL • Peripheral T-cell lymphoma NOS • Nodal PTCL with TFH • Angioimmunoblastic lymphoma • Follicular T-cell lymphoma • Anaplastic largecell lymphoma (ALK+/ALK/breast implant) EXTRANODAL • Extranodaln NK/T cell • Hepatosplenic • Entheropathy associated T-cell lymphoma • Mycosis fungoides • Sezary syndrom • Primary cutaneous CD30+ Swerdlow et al Blood 2016 348 2nd Summer School in medical oncology Standards and perspectives in the systemic treatment of Hodgkin lymphoma Urska Rugelj, MD Ljubljana, 10.9.2021 Conclusions • Incidence in EU 2.3/100.000, most often affects young adults • Multiple signaling pathways/transcription factors deregulated activities in RS cells involved in pathogenesis • 5-FDG PET-CT scan an important part of diagnostic and treatment modifying decisions • Type of treatment depends on clinical stage and risk factors • Combined approach of treatment in early stages, chemotherapy-based treatment for advance disease • Overall >80% achieve long remission – are cured • Standard care of relapse in young fit patient is high-dose ChT + AutoSCT • In relapse settings already approved use of some novel drugs as antibody-drug conjugates, anti PD-1 antibodies, others as small molecule inhibitors, cellular therapies in study settings 349 PRINCIPLES OF RADIOTHERAPY INLYMPHOMAS Assist. Prof. PhD Lorna Zadravec Zaletel, MD 2nd Summer School in medical oncology, 2021 Conclusions • malignant lymphomas are radiosensitive. • RT may be a stand-alone treatment for some indolent NHL and for low stage HL- type lymphocyte predominance . • in indolent NHL of higher stages, RT is reserved for treating the rest of the disease after CT or low-dose RT of larger infiltrates in order to delay CT. • in aggressive lymphomas, radiation associated with systemic treatment. is always • life-long follow-up of late sequelae of treatment is of vital importance. 350 SAMO ZA STROKOVNO JAVNOST ZAUSTAVITE NAPREDOVANJE BOLEZNI IN PODALJŠAJTE PREŽIVETJE Pri bolnikih z mHSPC, zdravljenje samo z ADT ni dovolj. ZDRAVILO ERLEADA® JE SEDAJ ODOBRENO TUDI ZA ZDRAVLJENJE BOLNIKOV S HORMONSKO OBČUTLJIVIM, METASTATSKIM RAKOM PROSTATE (mHSPC).¹ Zgodnja uporaba zdravila ERLEADA+ADT v primerjavi z ADT pomembno podaljša preživetje bolnikov in zmanjša tveganje za napredovanje bolezni, hkrati pa prihrani druge oblike zdravljenja za kasnejše stadije bolezni.1-3 ▼ Za to zdravilo se izvaja dodatno spremljanje varnosti. Tako bodo hitreje na voljo nove informacije o njegovi varnosti. Zdravstvene delavce naprošamo, da poročajo o katerem koli domnevnem neželenem učinku zdravila. Glejte poglavje 4.8 povzetka glavnih značilnosti zdravila, kako poročati o neželenih učinkih. Ime zdravila: Erleada 60 mg filmsko obložene tablete. Kakovostna in količinska sestava: 60 mg apalutamida; pomožne snovi: brezvodni koloidni silicijev dioksid, premreženi natrijev karmelozat, hipromeloza acetat sukcinat, magnezijev stearat, mikrokristalna celuloza, mikrokristalna celuloza (silicifirana), črni in rumeni železov dioksid, makrogol, polivinilalkohol (delno hidroliziran), smukec, titanov dioksid. Indikacije: Zdravljenje odraslih moških z nemetastatskim, na kastracijo odpornim rakom prostate (nmCRPC), pri katerih obstaja veliko tveganje za razvoj metastatske bolezni. Za zdravljenje odraslih moških s hormonsko občutljivim metastatskim rakom prostate (mHSCP) v kombinaciji z zdravljenjem z odtegnitvijo androgenov. Odmerjanje in način uporabe: Priporočeni odmerek je 240 mg (štiri 60‑miligramske tablete) v enkratnem peroralnem odmerku na dan. Med zdravljenjem je treba pri bolnikih, ki niso bili kirurško kastrirani, nadaljevati medicinsko kastracijo z analogom gonadoliberina. V primeru izpuščenega odmerka je treba zdravilo vzeti čimprej še isti dan, naslednji dan pa naj odmerjanje nadaljuje po običajnem razporedu. Dodatnih tablet za nadomestitev pozabljenega odmerka se ne sme vzeti. Če se pri bolniku pojavijo toksični učinki ≥ 3. stopnje ali nesprejemljivi neželeni učinki, je treba uporabo zdravila prekiniti začasno in ne dokončno, dokler se simptomi ne izboljšajo na ≤ 1. stopnjo oziroma na začetno stopnjo, nato pa z zdravljenjem nadaljevati z enakim ali manjšim odmerkom (180 mg ali 120 mg), če je potrebno. Starejšim bolnikom, bolnikom z blago do zmerno okvaro ledvic ali jeter odmerka ni treba prilagajati. Pri bolnikih s hudo okvaro ledvic je potrebna previdnost, pri bolnikih s hudo okvaro jeter pa uporaba ni priporočljiva. Tablete je treba pogoltniti cele in se jih lahko jemlje s hrano ali brez nje. Apalutamid ni namenjen za uporabo pri pediatrični populaciji. Kontraindikacije: Preobčutljivost na učinkovino ali katero koli pomožno snov, nosečnice in ženske, ki bi lahko zanosile. Posebna opozorila in previdnostni ukrepi: Uporaba zdravila ni priporočljiva pri bolnikih z anamnezo konvulzij ali drugimi predispozicijskimi dejavniki, med drugim tudi pri bolnikih s poškodbo možganov, nedavno kapjo (v zadnjem letu), pri bolnikih s primarnimi možganskimi tumorji ali metastazami v možganih. Pri bolnikih, ki so prejemali apalutamid je prišlo do padcev in zlomov, zato je treba pred uvedbo zdravljenja pri bolnikih oceniti tveganje za zlome in padce, bolnike pa spremljati po ustaljenih smernicah in premisliti o uporabi učinkovin, ki delujejo na kosti. Bolnike je treba spremljati tudi glede znakov in simptomov ishemične bolezni srca in ishemičnih možganskožilnih bolezni ter optimatizirati obvladovanje dejavnikov tveganja, kot so hipertenzija, diabetes ali dislipidemija, skladno s standardno oskrbo. Sočasni uporabi apalutamida z zdravili, ki so občutljivi substrati več presnovnih encimov ali prenašalcev, se je načeloma treba izogibati, če je terapevtski učinek teh zdravil za bolnika zelo pomemben in njihovega odmerjanja ni mogoče enostavno prilagajati na osnovi spremljanja učinkovitosti ali koncentracij v plazmi. Sočasni uporabi z varfarinom ali kumarinskimi antikoagulansi se je treba izogibati. Če se predpiše apalutamid, je treba pri bolnikih s klinično pomembnimi boleznimi srca in ožilja spremljati dejavnike tveganja kot so hiperholesterolemija, hipertrigliceridemija ali druge srčno presnovne bolezni. Zdravljenje z odtegnitvijo androgenov lahko Janssen, farmacevtski del Johnson & Johnson d.o.o., Šmartinska cesta 53, 1000 Ljubljana, tel: 01 401 18 00, e-mail: info@janssen-slovenia.si podaljša interval QT. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij: Apalutamid je induktor encimov in prenašalcev in lahko povzroči povečan obseg odstranjevanja številnih pogosto uporabljanih zdravil. Pri sočasnem odmerjanju tega zdravila s katerim od močnih zaviralcev CYP2C8 ali močnih zaviralcev CYP3A4 začetnega odmerka ni treba prilagajati, premisliti pa velja o zmanjšanju odmerka zdravila Erleada na osnovi prenašanja zdravila. Ni pričakovati, da bi induktorji CYP3A4 ali CYP2C8 klinično pomembno vplivali na farmakokinetiko apalutamida in aktivnih frakcij. . Pri sočasni uporabi s substrati CYP2B6 je treba spremljati neželene učinke in oceniti izgubo učinka substrata ter za zagotovitev optimalnih plazemskih koncentracij morda prilagoditi odmerek substrata. Sočasna uporaba z zdravili, ki se primarno presnavljajo s CYP3A4 (kot so darunavir, felodipin, midazolam in simvastatin), s CYP2C19 (kot sta diazepam in omeprazol) ali s CYP2C9 (kot sta varfarin in fenitoin), lahko povzroči zmanjšanje izpostavljenosti tem zdravilom. Pri sočasni uporabi s substrati UDP‑glukuronil transferaze je potrebna previdnost. Pri sočasni uporabi s substrati P‑gp, BCRP ali OATP1B1 je potrebna ocena obsega zmanjšanja učinka ter za zagotovitev optimalnih plazemskih koncentracij morda prilagoditi odmerek substrata. Ni mogoče izključiti možnosti, da apalutamid in njegov N‑desmetil presnovek zavirata prenašalce OCT2,OAT3 in MATE. Pri preiskovancih z mHSPC, ki so prejemali levprorelinijev acetat (analog GnRH), sočasna uporaba apalutamida ni bistveno vplivala na izpostavljenost leuprolidu v stanju dinamičnega ravnovesja. Skrbna presoja je potrebna tudi pri sočasni uporabi z zdravili, za katera je ugotovljeno, da podaljšujejo interval QT, oziroma z zdravili, ki lahko izzovejo Torsades de pointes. Plodnost, nosečnost in dojenje: Ni znano, ali so apalutamid ali njegovi presnovki prisotni v spermi, zato lahko to zdravilo škoduje plodu v razvoju. Bolniki, ki imajo spolne odnose z žensko v rodni dobi, morajo med zdravljenjem in še 3 mesece po zadnjem odmerku zdravila Erleada uporabljati kondome skupaj s še katero od drugih visoko učinkovitih metod kontracepcije. Zdravilo je kontraindicirano pri nosečnicah in ženskah , ki bi lahko zanosile in se ne sme uporabljati med dojenjem. Neželeni učinki: Hipotiroidizem, zmanjšan apetit, hiperholesterolemija, hipertrigliceridemija, disgevzija, ishemične možganskožilne bolezni, konvulzije, ishemična bolezen srca, podaljšanje intervala QT, vročinski oblivi, hipertenzija, driska, kožni izpuščaj, srbenje, alopecija, TEN, zlomi, artralgija, mišični krči, utrujenost, zmanjšanje telesne mase, padci. Za popoln seznam neželenih učinkov glejte Povzetek glavnih značilnosti zdravila. Imetnik DzP: Janssen-Cilag International NV, Turnhoutseweg 30, 2340 Beerse, Belgija Predstavnik imetnika DzP v Sloveniji: Johnson & Johnson d.o.o., Šmartinska cesta 53, Ljubljana. Režim izdajanja zdravila: Rp/Spec. Datum zadnje revizije besedila: 10. junij 2021 Povzetek glavnih značilnosti zdravila s podrobnejšimi informacijami o zdravilu je dostopen pri predstavniku imetnika dovoljenja za promet. Viri: 1. Povzetek glavnih značilnosti zdravila ERLEADA® (apalutamid). 2. Chi KN, et al. N Engl J Med. 2019;81(1):13–24 3. Chi KN, et al. N Engl J Med. 2019;81(1):13–24. Supplementary information. (apalutamid) tablete CP-198062/020921 Skrajšan povzetek glavnih značilnosti zdravila ERLEADA® SAMO ZA STROKOVNO JAVNOST Prevzemite nadzor nad KLL, MCL in WM z zdravilom IMBRUVICA®1-6 ENA TABLETA ENKRAT NA DAN DOMA Dolgotrajno izboljšanje preživetja po 8 letih spremljanja.2-3 Poznan in obvladljiv varnostni profil.2-6 Enostavno, enkrat dnevno jemanje zdravila na domu.1 tablete KLL=kronična limfocitna levkemija; MCL=limfom plaščnih celic; WM=Waldenströmova makroglobulinemija. SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA Ime zdravila: IMBRUVICA 140 mg/280 mg/420 mg/560 mg filmsko obložene tablete Kakovostna in količinska sestava: 140/280/420 ali 560 mg ibrutiniba, brezvodni koloidni silicijev dioksid, premrežen natrijev karmelozat, laktoza monohidrat, magnezijev stearat, mikrokristalna celuloza, natrijev lavrilsulfat (E487), povidon; filmska obloga: makrogol, polivinilalkohol, smukec, titanov dioksid (E171), črni železov oksid (E172), rumeni železov oksid (E172 – 140, 420 in 560 mg), rdeči železov oksid (E172 – 280 in 560 mg) Indikacije: Kot samostojno zdravilo: zdravljenje odraslih bolnikov s ponovitvijo limfoma plaščnih celic (MCL) ali z na zdravljenje neodzivno obliko te bolezni, zdravljenje odraslih bolnikov z Waldenstromovo makroglobulinemijo (WM - Waldenström’s macroglobulinaemia), ki so predhodno prejeli vsaj eno vrsto zdravljenja oziroma v prvi liniji pri bolnikih, ki niso primerni za kemoimunoterapijo. Kot samostojno zdravilo ali v kombinaciji z obinutuzumabom za zdravljenje odraslih bolnikov s predhodno neozdravljivo kronično limfocitno levkemijo (KLL)/z bendamustinom in rituksimabom za zdravljenje odraslih bolnikov s KLL, ki so predhodno prejeli vsaj eno vrsto zdravljenja; samostojno zdravilo indicirano. V kombinaciji z rituksimabom za zdravljenje odraslih bolnikov z WM. Odmerjanje: Zdravilo je treba jemati peroralno enkrat na dan s kozarcem vode, in sicer vsak dan ob približno istem času. Tablete je treba pogoltniti cele z vodo. Tablet se ne sme drobiti ali žvečiti. Zdravljenje mora uvesti in nadzorovati zdravnik, ki ima izkušnje z uporabo onkoloških zdravil. MCL: priporočeni odmerek je 560 mg enkrat na dan. KLL (samostojno ali v kombinaciji) in WM: priporočeni odmerek je 420 mg enkrat na dan. Z zdravljenjem je treba nadaljevati do napredovanja bolezni oz. dokler bolnik zdravilo prenaša. Pri odmerjanju tega zdravila v kombinaciji z zdravilom, ki je usmerjeno proti CD20, je priporočljivo vzeti ibrutinib pred zdravilom, ki je usmerjeno proti CD20, kadar ju je treba vzeti na isti dan. Podrobna navodila za odmerjanje pri posebnih skupinah bolnikov in za prilagajanje odmerkov v primeru sočasne uporabe z zmernimi in močnimi zaviralci CYP3A4 in ob pojavu hematološke toksičnosti so navedena v Povzetku glavnih značilnosti zdravila. Če bolnik izpusti odmerek, ga lahko vzame čimprej istega dne, naslednjega dne pa spet začne z odmerjanjem po običajnem razporedu. Bolnik naj ne jemlje dodatnih tablet, da bi nadomestil pozabljeni odmerek. Kontraindikacije: Preobčutljivost na učinkovino ali katero koli pomožno snov, sočasna uporaba pripravkov rastlinskega izvora s šentanževko (Hypericum perforatum). Posebna opozorila in previdnostni ukrepi: Poročali so o krvavitvah s trombocitopenijo ali brez nje (vključno z manjšimi hemoragičnimi dogodki (podplutbe, krvavitev iz nosu, petehije) ter večjimi krvavitvami (gastrointestinalna in intrakranialna krvavitev, hematurija)). Sočasno se ne sme jemati varfarina in drugih antagonistov vitamina K. Izogibati se je treba prehranskim dopolnilom (npr. pripravki ribjega olja ali vitamina E). Pri sočasnem zdravljenju z antikoagulanti je potrebna posebna previdnost. Zdravila se ne sme jemati najmanj 3 do 7 dni (odvisno od vrste kirurškega posega in tveganja za krvavitev) pred kirurškim posegom in po njem. Poročali so o primerih levkostaze. Po prekinitvi zdravljenja z ibrutinibom so poročali o primerih rupture vranice. Veliko število cirkulirajočih limfocitov (> 400.000/mikroliter) lahko pomeni povečano tveganje; treba je razmisliti o začasni prekinitvi jemanja zdravila, bolnika skrbno spremljati in po potrebi uvesti podporne ukrepe vključno s hidracijo in/ali citoredukcijo. Opažali so okužbe; bolnike je treba spremljati glede morebitnega pojava zvišane telesne temperature, nenormalnih izvidov preiskav delovanja jeter, nevtropenije in okužbe ter po potrebi uvesti ustrezno antimikrobno zdravljenje. Pri bolnikih s povečanim tveganjem za oportunistične okužbe razmislite o standardnih ukrepih za njihovo preprečevanje. Po uporabi ibrutiniba so poročali so o primerih invazivnih glivičnih okužb, vključno s primeri aspergiloze, kriptokokoze in okužbe s Pneumocystis jiroveci. Pri uporabi ibrutiniba ob predhodni ali sočasni uporabi imunosupresivnega zdravljenja so poročali o PML, vključno s smrtnimi primeri. PML je treba upoštevati v diferencialni diagnozi pri bolnikih z novimi ali s poslabšanjem obstoječih nevroloških, kognitivnih ali vedenjskih znakov ali simptomov. Če obstaja sum za PML, je treba opraviti diagnostične preiskave in zdravljenje prekiniti dokler ni izključena. Pri bolnikih, zdravljenih z ibrutinibom so se pojavili primeri hepatotoksičnosti, reaktivacije virusa hepatitisa B in primeri hepatitisa E, ki so lahko kronične narave, prišlo je tudi do odpovedi jeter, vključno s smrtnimi izidi. Pred uvedbo zdravila je treba preveriti delovanje jeter in prisotnost virusa hepatitisa. Poročali so tudi o citopenijah, zato je treba enkrat mesečno določati celotno krvno sliko. Pri bolnikih, ki so jemali ibrutinib so poročali tudi o primerih atrijske fibrilacije, atrijske undulacije, ventrikularne tahiaritmije in srčnega popuščanja. Zaradi možnosti pojava srčnih bolezni, vključno s srčnimi aritmijami in srčnim popuščanjem je treba vse bolnike občasno klinično pregledati. Pri bolnikih, pri katerih so se pojavili simptomi in/ali znaki ventrikularne tahiaritmije je treba zdravljenje s tem zdravilom začasno prekiniti, pred ponovno uvedbo je treba temeljito oceniti klinično razmerje med koristjo in tveganjem. Bolnike, pri katerih se pojavijo simptomi aritmije ali se na novo pojavi zadihanost, omotica ali omedlevica, je treba klinično pregledati in jim po potrebi posneti EKG. Pri bolnikih z obstoječo atrijsko fibrilacijo, ki potrebujejo zdravljenje z antikoagulanti, je treba razmisliti o drugih možnostih zdravljenja. Če se atrijska fibrilacija pojavi med zdravljenjem, je treba temeljito oceniti tveganje za trombembolične bolezni. Pri bolnikih z velikim tveganjem in kadar druge možnosti zdravljenja niso primerne, je treba razmisliti o skrbno nadzorovanem zdravljenju z antikoagulanti Bolnike je treba med zdravljenjem skrbno spremljati glede znakov in simptomov srčnega popuščanja. Pri bolnikih, ki so jemali to zdravilo so poročali o primerih ILD. Bolnike spremljajte glede pljučnih simptomov ILD. Če se simptomi pojavijo, je treba zdravljenje prekiniti in ILD ustrezno zdraviti. Če simptomi vztrajajo je treba oceniti tveganje in korist zdravljenja in upoštevati smernice za prilagoditev odmerjanja. Pri bolnikih, ki so prejemali ibrutinib so poročali o primerih cerebrovaskularnega insulta, prehodnega ishemičnega napada in ishemične možganske kapi s sočasno atrijsko fibrilacijo in/ali hipertenzijo ali brez njiju. Med primeri, ki so bili poročani z zakasnitvijo, je od začetka zdravljenja do pojava ishemičnih žilnih bolezni osrednjega Izkušnje, na katere se lahko zanesete živčevja večinoma minilo nekaj mesecev. Bolnike z večjo maso tumorja pred začetkom zdravljenja je treba skrbno spremljati zaradi večjega tveganja za pojav sindroma razpada tumorja. Med zdravljenjem je treba bolnike spremljati glede morebitnega pojava nemelanomskega kožnega raka. Bolnikom, ki prejemajo ibrutinib, je treba med celotnim potekom zdravljenja redno meriti krvni tlak in jim po potrebi uvesti ali prilagoditi odmerjanje antihipertenzivnih zdravil. Poročali so tudi o primerih hemofagocitne limfohistiocitoze, vključno s smrtnimi primeri. Pri sočasni uporabi z zmernimi/močnimi zaviralci CYP3A4 lahko pride do povečane izpostavljenosti ibrutinibu in večjega tveganja za pojav toksičnosti, pri sočasni uporabi z indukdorji CYP3A4 pa do zmanjšane izpostavljenosti in tveganja za pomanjkanje učinkovitosti. Zato se je treba sočasni uporabi z močnimi zaviralci CYP3A4 in močnimi ali zmernimi induktorji CYP3A4 izogibati. O sočasni uporabi lahko razmislite samo, kadar pričakovane koristi nedvoumno presegajo morebitno tveganje. Pri bolnikih, ki morajo jemati zaviralce CYP3A4, je treba skrbno spremljati morebitne znake toksičnega delovanja zdravila; pri tistih, ki jemljejo induktorje CYP3A4 pa znake pomanjkanja učinkovitosti. Bolniki z redko dedno intoleranco za galaktozo, odsotnostjo encima laktaze ali malabsorpcijo glukoze/galaktoze ne smejo jemati tega zdravila. Ena filmsko obložena tableta vsebuje manj kot 1 mmol natrija (23 mg) kar v bistvu pomeni ‘brez natrija’. Interakcije: Sočasna uporaba z zdravili, ki močno/zmerno zavirajo CYP3A4, lahko poveča izpostavljenost ibrutinibu, zato se je treba uporabi močnih zaviralcev CYP3A4 izogibati. Če mora bolnik jemati katerega od močnih/zmernih zaviralcev CYP3A4, je treba odmerek zdravila Imbruvica prilagoditi (glejte Povzetek glavnih značilnosti zdravila), bolnike pa skrbno spremljati. V kombinaciji s šibkimi zaviralci prilagajanje odmerjanja ni potrebno. Bolnike je treba skrbno spremljati in po potrebi upoštevati smernice za prilagajanje odmerka. Med zdravljenjem se je treba izogibati uživanju grenivk in seviljskih pomaranč, ker vsebujejo zmerne zaviralce CYP3A4. Sočasna uporaba zdravila Imbruvica z induktorji CYP3A4 lahko zmanjša koncentracijo ibrutiniba v plazmi. Razmisliti velja o uporabi drugih učinkovin, ki v manjši meri inducirajo CYP3A4. Če je potrebna uporaba močnega ali zmernega induktorja CYP3A4 in pričakovana korist presega morebitno tveganje, je treba bolnika skrbno spremljati glede znakov pomanjkanja učinkovitosti. Zdravilo se lahko sočasno uporablja z blagimi induktorji, vendar je treba bolnike skrbno spremljati glede znakov pomanjkanja učinkovitosti. Topnost ibrutiniba je odvisna od pH, zato lahko zdravila, ki zvečajo pH v želodcu (npr. zaviralci protonske črpalke), zmanjšajo izpostavljenost ibrutinibu. In vitro ibrutinib zavira P‑glikoprotein in BCRP, zato je treba substrate P‑glikoproteina in BCRP, ki imajo ozko peroralno terapevtsko okno (npr. digoksin, metotreksat) jemati najmanj 6 ur pred oz. najmanj 6 ur po odmerjanju zdravila Imbruvica. Ibrutinib lahko zavira tudi BCRP v jetrih in zveča izpostavljenost zdravilom, katerih izločanje skozi jetra je povezano z BCRP (rosuvastatin). V študiji medsebojnega delovanja z drugimi zdravili pri bolnikih z malignomi celic B, ibrutinib v enkratnem, 560 mg odmerku ni klinično pomembno vplival na izpostavljenost substratu CYP3A4 midazolamu. V isti študiji, 2 tedensko zdravljenje z ibrutinibom v odmerku 560 mg na dan, ni klinično pomembno vplivalo na farmakokinetiko oralnih kontraceptivov (etiniletradiol in levonorgestrel), substrata CYP3A4 midazolama ali substrata CYP2B6 bupropiona. Nosečnost, dojenje in plodnost: Ženske v rodni dobi morajo med zdravljenjem in še tri mesece po zaključku zdravljenja uporabljati zelo učinkovito metodo kontracepcije. Zdravila ne smete uporabljati pri nosečnicah. Med zdravljenjem je treba prenehati z dojenjem. Podatkov o vplivu ibrutiniba na plodnost pri ljudeh ni na voljo. Vpliv na sposobnost vožnje in upravljanja strojev: Zdravilo ima blag vpliv na sposobnost vožnje in upravljanja strojev. Pri nekaterih bolnikih so poročali o utrujenosti, omotičnosti in asteniji, kar je treba upoštevati pri presoji bolnikove sposobnosti za vožnjo in upravljanje strojev. Neželeni učinki: pljučnica, okužba zgornjih dihal, sinusitis, sepsa, kriptokokna/pnevmocistična okužba, okužba z aspergilusom, reaktivacija hepatitisa B, nemelanomski kožni raki, okužba sečil, okužba kože, nevtropenija, trombocitopenija, limfocitoza, anemija, febrilna nevtropenija, levkocitoza, sindrom levkostaze, ILD, dehidracija, hiperurikemija, sindrom razpada tumorja, omotičnost, glavobol, periferna nevropatija, cerebrovaskularni insult, prehodni ishemični napad, ishemična možganska kap, zamegljen vid, očesna krvavitev, srčno popuščanje, atrijska fibrilacija, ventrikularna tahiaritmija, krvavitev, podplutba,petehije, subduralni hematom, krvavitev iz nosu, diareja, bruhanje, stomatitis, navzea, obstipacija, suha usta, odpoved jeter, izpuščaj, angioedem, panikulitis, nevtrofilne dermatoze, urtikarija, eritem, lomljenje nohtov, Stevens-Johnsonov sindrom, artralgija, mišičnoskeletne bolečine, zvišana telesna temperatura, periferni edem, zvišanje kreatinina v krvi (vsi NU so opisani v povzetku glavnih značilnosti zdravila) Način in režim izdajanja zdravila: Rp/Spec. Imetnik DzP: Janssen-Cilag International NV, Turnhoutseweg 30, 2340 Beerse, Belgija Predstavnik Imetnika DzP v Sloveniji: Johnson & Johnson d.o.o., Šmartinska cesta 53, 1000 Ljubljana Datum zadnje revizije besedila: 20. 08. 2021 Povzetek glavnih značilnosti zdravila s podrobnejšimi informacijami o zdravilu je dostopen pri predstavniku imetnika dovoljenja za promet. Viri: 1. Povzetek glavnih značilnosti zdravila IMBRUVICA® (ibrutinib). 2. Byrd JC et al. Clin Cancer Res. 2020 Aug 1;26(15):3918-3927. 3. Burger JA, et al. Leukemia 2020;34(3):787-798. 4. Munir T, et al. Am J Hematol 2019;94(12):1353-1363. 5. Treon SP, et al. Haem Onc 2019;37(S2):184-185. 6. O’Brien S, et al. Clin Lymphoma Myeloma Leuk 2018;18(10):648-657. Janssen, farmacevtski del Johnson & Johnson d.o.o. Šmartinska 53, 1000 Ljubljana, Slovenia, Tel: 01 401 18 00, e-mail: info@janssen-slovenia.si, www.janssen.com/slovenia CP-214010/020921 (pembrolizumab, MSD) Kolorektalni rak1 Nedrobnocelični pljučni rak1 Rak ledvičnih celic1 Melanom1 Referenca: 1. Keytruda EU SmPC SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA Pred predpisovanjem, prosimo, preberite celoten Povzetek glavnih značilnosti zdravila! Ime zdravila: KEYTRUDA 25 mg/ml koncentrat za raztopino za infundiranje vsebuje pembrolizumab. Terapevtske indikacije: Zdravilo KEYTRUDA je kot samostojno zdravljenje indicirano za zdravljenje: napredovalega (neoperabilnega ali metastatskega) melanoma pri odraslih; za adjuvantno zdravljenje odraslih z melanomom v stadiju III, ki se je razširil na bezgavke, po popolni kirurški odstranitvi; metastatskega nedrobnoceličnega pljučnega raka (NSCLC) v prvi liniji zdravljenja pri odraslih, ki imajo tumorje z ≥ 50 % izraženostjo PD-L1 (TPS) in brez pozitivnih tumorskih mutacij EGFR ali ALK; lokalno napredovalega ali metastatskega NSCLC pri odraslih, ki imajo tumorje z ≥ 1 % izraženostjo PD-L1 (TPS) in so bili predhodno zdravljeni z vsaj eno shemo kemoterapije, bolniki s pozitivnimi tumorskimi mutacijami EGFR ali ALK so pred prejemom zdravila KEYTRUDA morali prejeti tudi tarčno zdravljenje; odraslih in pediatričnih bolnikov, starih 3 leta ali več, s ponovljenim ali neodzivnim klasičnim Hodgkinovim limfomom (cHL), pri katerih avtologna presaditev matičnih celic (ASCT) ni bila uspešna, ali po najmanj dveh predhodnih zdravljenjih kadar ASCT ne pride v poštev kot možnost zdravljenja; lokalno napredovalega ali metastatskega urotelijskega raka pri odraslih, predhodno zdravljenih s kemoterapijo, ki je vključevala platino; lokalno napredovalega ali metastatskega urotelijskega raka pri odraslih, ki niso primerni za zdravljenje s kemoterapijo, ki vsebuje cisplatin in imajo tumorje z izraženostjo PD-L1 ≥ 10, ocenjeno s kombinirano pozitivno oceno (CPS); ponovljenega ali metastatskega ploščatoceličnega raka glave in vratu (HNSCC) pri odraslih, ki imajo tumorje z ≥ 50 % izraženostjo PD-L1 (TPS), in pri katerih je bolezen napredovala med zdravljenjem ali po zdravljenju s kemoterapijo, ki je vključevala platino in za prvo linijo zdravljenja metastatskega kolorektalnega raka z visoko mikrosatelitsko nestabilnostjo (MSI-H – microsatellite instability-high) ali s pomanjkljivim popravljanjem neujemanja pri podvojevanju DNA (dMMR - mismatch repair deficient) pri odraslih. Zdravilo KEYTRUDA je kot samostojno zdravljenje ali v kombinaciji s kemoterapijo s platino in 5-fluorouracilom (5-FU) indicirano za prvo linijo zdravljenja metastatskega ali neoperabilnega ponovljenega ploščatoceličnega raka glave in vratu pri odraslih, ki imajo tumorje z izraženostjo PD-L1 s CPS ≥ 1. Zdravilo KEYTRUDA je v kombinaciji s pemetreksedom in kemoterapijo na osnovi platine indicirano za prvo linijo zdravljenja metastatskega neploščatoceličnega NSCLC pri odraslih, pri katerih tumorji nimajo pozitivnih mutacij EGFR ali ALK; v kombinaciji s karboplatinom in bodisi paklitakselom bodisi nab-paklitakselom je indicirano za prvo linijo zdravljenja metastatskega ploščatoceličnega NSCLC pri odraslih; v kombinaciji z aksitinibom je indicirano za prvo linijo zdravljenja napredovalega raka ledvičnih celic (RCC) pri odraslih; v kombinaciji s kemoterapijo s platino in fluoropirimidinom indicirano za prvo linijo zdravljenja lokalno napredovalega neoperabilnega ali metastatskega raka požiralnika ali HER-2 negativnega adenokarcinoma gastroezofagealnega prehoda pri odraslih, ki imajo tumorje z izraženostjo PD-L1 s CPS ≥ 10. Odmerjanje in način uporabe: Testiranje PD-L1: Če je navedeno v indikaciji, je treba izbiro bolnika za zdravljenje z zdravilom KEYTRUDA na podlagi izraženosti PD-L1 tumorja potrditi z validirano preiskavo. Testiranje MSI-H/ dMMR pri bolnikih s CRC: Za samostojno zdravljenje z zdravilom KEYTRUDA je priporočljivo opraviti testiranje MSI-H/dMMR statusa tumorja z validirano preiskavo, da se izbere bolnike s CRC. Odmerjanje: Priporočeni odmerek zdravila KEYTRUDA pri odraslih je bodisi 200 mg na 3 tedne ali 400 mg na 6 tednov, apliciran z intravensko infuzijo v 30 minutah. Priporočeni odmerek zdravila KEYTRUDA za samostojno zdravljenje pri pediatričnih bolnikih s cHL, starih 3 leta ali več, je 2 mg/kg telesne mase (do največ 200 mg) na 3 tedne, apliciran z intravensko infuzijo v 30 minutah. Za uporabo v kombinaciji glejte povzetke glavnih značilnosti sočasno uporabljenih zdravil. Če se uporablja kot del kombiniranega zdravljenja skupaj z intravensko kemoterapijo, je treba zdravilo KEYTRUDA aplicirati prvo. Bolnike je treba zdraviti do napredovanja bolezni ali nesprejemljivih toksičnih učinkov. Pri adjuvantnem zdravljenju melanoma je treba zdravilo uporabljati do ponovitve bolezni, pojava nesprejemljivih toksičnih učinkov oziroma mora zdravljenje trajati do enega leta. Če je aksitinib uporabljen v kombinaciji s pembrolizumabom, se lahko razmisli o povečanju odmerka aksitiniba nad začetnih 5 mg v presledkih šest tednov ali več. Pri bolnikih starih ≥ 65 let, bolnikih z blago do zmerno okvaro ledvic, bolnikih z blago okvaro jeter prilagoditev odmerka ni potrebna. Odložitev odmerka ali ukinitev zdravljenja: Zmanjšanje odmerka zdravila KEYTRUDA ni priporočljivo. Za obvladovanje neželenih učinkov je treba uporabo zdravila KEYTRUDA zadržati ali ukiniti, prosimo, glejte celoten Povzetek glavnih značilnosti zdravila. Kontraindikacije: Preobčutljivost na učinkovino ali katero koli pomožno snov. Povzetek posebnih opozoril, previdnostnih ukrepov, interakcij in neželenih učinkov: Imunsko pogojeni neželeni učinki (pnevmonitis, kolitis, hepatitis, nefritis, endokrinopatije, neželeni učinki Ploščatocelični karcinom glave in vratu1 Hodgkinov limfom1 Rak požiralnika1 Urotelijski karcinom1 na kožo in drugi): Pri bolnikih, ki so prejemali pembrolizumab, so se pojavili imunsko pogojeni neželeni učinki, vključno s hudimi in smrtnimi primeri. Večina imunsko pogojenih neželenih učinkov, ki so se pojavili med zdravljenjem s pembrolizumabom, je bila reverzibilnih in so jih obvladali s prekinitvami uporabe pembrolizumaba, uporabo kortikosteroidov in/ali podporno oskrbo. Pojavijo se lahko tudi po zadnjem odmerku pembrolizumaba in hkrati prizadanejo več organskih sistemov. V primeru suma na imunsko pogojene neželene učinke je treba poskrbeti za ustrezno oceno za potrditev etiologije oziroma izključitev drugih vzrokov. Glede na izrazitost neželenega učinka je treba zadržati uporabo pembrolizumaba in uporabiti kortikosteroide – za natančna navodila, prosimo, glejte Povzetek glavnih značilnosti zdravila Keytruda. Zdravljenje s pembrolizumabom lahko poveča tveganje za zavrnitev pri prejemnikih presadkov čvrstih organov. Pri bolnikih, ki so prejemali pembrolizumab, so poročali o hudih z infuzijo povezanih reakcijah, vključno s preobčutljivostjo in anafilaksijo. Pembrolizumab se iz obtoka odstrani s katabolizmom, zato presnovnih medsebojnih delovanj zdravil ni pričakovati. Uporabi sistemskih kortikosteroidov ali imunosupresivov pred uvedbo pembrolizumaba se je treba izogibati, ker lahko vplivajo na farmakodinamično aktivnost in učinkovitost pembrolizumaba. Vendar pa je kortikosteroide ali druge imunosupresive mogoče uporabiti za zdravljenje imunsko pogojenih neželenih učinkov. Kortikosteroide je mogoče uporabiti tudi kot premedikacijo, če je pembrolizumab uporabljen v kombinaciji s kemoterapijo, kot antiemetično profilakso in/ali za ublažitev neželenih učinkov, povezanih s kemoterapijo. Ženske v rodni dobi morajo med zdravljenjem s pembrolizumabom in vsaj še 4 mesece po zadnjem odmerku pembrolizumaba uporabljati učinkovito kontracepcijo, med nosečnostjo in dojenjem se ga ne sme uporabljati. Varnost pembrolizumaba pri samostojnem zdravljenju so v kliničnih študijah ocenili pri 6.185 bolnikih z napredovalim melanomom, kirurško odstranjenim melanomom v stadiju III (adjuvantno zdravljenje), NSCLC, cHL, urotelijskim rakom, HNSCC ali CRC s štirimi odmerki (2 mg/kg telesne mase na 3 tedne, 200 mg na 3 tedne in 10 mg/ kg telesne mase na 2 ali 3 tedne). V tej populaciji bolnikov je mediani čas opazovanja znašal 7,6 mesece (v razponu od 1 dneva do 47 mesecev), najpogostejši neželeni učinki zdravljenja s pembrolizumabom so bili utrujenost (32 %), navzea (21 %) in diareja (21 %). Večina poročanih neželenih učinkov pri samostojnem zdravljenju je bila po izrazitosti 1. ali 2. stopnje. Najresnejši neželeni učinki so bili imunsko pogojeni neželeni učinki in hude z infuzijo povezane reakcije. Varnost pembrolizumaba pri kombiniranem zdravljenju s kemoterapijo so ocenili pri 1.437 bolnikih NSCLC, HNSCC ali rakom požiralnika, ki so v kliničnih študijah prejemali pembrolizumab v odmerkih 200 mg, 2 mg/kg telesne mase ali 10 mg/kg telesne mase na vsake 3 tedne. V tej populaciji bolnikov so bili najpogostejši neželeni učinki naslednji: navzea (55 %), anemija (51 %), utrujenost (39 %), zaprtost (37 %), zmanjšanje apetita (34 %), diareja (33 %), nevtropenija (29 %) in bruhanje (28 %). Pojavnost neželenih učinkov 3. do 5. stopnje je pri bolnikih z NSCLC pri kombiniranem zdravljenju s pembrolizumabom znašala 67 % in pri zdravljenju samo s kemoterapijo 66 %, pri bolnikih s HNSCC pri kombiniranem zdravljenju s pembrolizumabom 85 % in pri zdravljenju s kemoterapijo v kombinaciji s cetuksimabom 84 %, ter pri bolnikih z rakom požiralnika pri kombiniranem zdravljenju s pembrolizumabom 86 % in pri zdravljenju samo s kemoterapijo 83 %. Varnost pembrolizumaba v kombinaciji z aksitinibom so ocenili v klinični študiji pri 429 bolnikih z napredovalim rakom ledvičnih celic, ki so prejemali 200 mg pembrolizumaba na 3 tedne in 5 mg aksitiniba dvakrat na dan. V tej populaciji bolnikov so bili najpogostejši neželeni učinki diareja (54 %), hipertenzija (45 %), utrujenost (38 %), hipotiroidizem (35 %), zmanjšan apetit (30 %), sindrom palmarnoplantarne eritrodisestezije (28 %), navzea (28 %), zvišanje vrednosti ALT (27 %), zvišanje vrednosti AST (26 %), disfonija (25 %), kašelj (21 %) in zaprtost (21 %). Pojavnost neželenih učinkov 3. do 5. stopnje je bila med kombiniranim zdravljenjem s pembrolizumabom 76 % in pri zdravljenju s sunitinibom samim 71 %. Za celoten seznam neželenih učinkov, prosimo, glejte celoten Povzetek glavnih značilnosti zdravila. Način in režim izdaje zdravila: H – Predpisovanje in izdaja zdravila je le na recept, zdravilo se uporablja samo v bolnišnicah. Imetnik dovoljenja za promet z zdravilom: Merck Sharp & Dohme B.V. , Waarderweg 39, 2031 BN Haarlem, Nizozemska. Merck Sharp & Dohme inovativna zdravila d.o.o., Ameriška ulica 2, 1000 Ljubljana, tel: +386 1/ 520 42 01, fax: +386 1/ 520 43 50; Pripravljeno v Sloveniji, julij 2021; SI-KEY-00304 EXP: 07/2023 Samo za strokovno javnost. H - Predpisovanje in izdaja zdravila je le na recept, zdravilo pa se uporablja samo v bolnišnicah. Pred predpisovanjem, prosimo, preberite celoten Povzetek glavnih značilnosti zdravila Keytruda, ki je na voljo pri naših strokovnih sodelavcih ali na lokalnem sedežu družbe. Zdravilo KEYTRUDA® (pembrolizumab, MSD) OMOGOČA VEČ ČASA Q6W - samo 9 infuzij letno* ODMERJANJE NA 6 TEDNOV: MANJ INFUZIJ ZA VAŠE BOLNIKE, VEČ ČASA ZA VAS! * Priporočeni odmerek zdravila KEYTRUDA pri odraslih je bodisi 200 mg na 3 tedne ali 400 mg na 6 tednov, apliciran z intravensko infuzijo v 30 minutah. Q3W = vsake 3 tedne; Q6W = vsakih 6 tednov. Referenca: 1. Keytruda EU SmPC SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA Pred predpisovanjem, prosimo, preberite celoten Povzetek glavnih značilnosti zdravila! Ime zdravila: KEYTRUDA 25 mg/ml koncentrat za raztopino za infundiranje vsebuje pembrolizumab. Terapevtske indikacije: Zdravilo KEYTRUDA je kot samostojno zdravljenje indicirano za zdravljenje: napredovalega (neoperabilnega ali metastatskega) melanoma pri odraslih; za adjuvantno zdravljenje odraslih z melanomom v stadiju III, ki se je razširil na bezgavke, po popolni kirurški odstranitvi; metastatskega nedrobnoceličnega pljučnega raka (NSCLC) v prvi liniji zdravljenja pri odraslih, ki imajo tumorje z ≥ 50 % izraženostjo PD-L1 (TPS) in brez pozitivnih tumorskih mutacij EGFR ali ALK; lokalno napredovalega ali metastatskega NSCLC pri odraslih, ki imajo tumorje z ≥ 1 % izraženostjo PD-L1 (TPS) in so bili predhodno zdravljeni z vsaj eno shemo kemoterapije, bolniki s pozitivnimi tumorskimi mutacijami EGFR ali ALK so pred prejemom zdravila KEYTRUDA morali prejeti tudi tarčno zdravljenje; odraslih in pediatričnih bolnikov, starih 3 leta ali več, s ponovljenim ali neodzivnim klasičnim Hodgkinovim limfomom (cHL), pri katerih avtologna presaditev matičnih celic (ASCT) ni bila uspešna, ali po najmanj dveh predhodnih zdravljenjih kadar ASCT ne pride v poštev kot možnost zdravljenja; lokalno napredovalega ali metastatskega urotelijskega raka pri odraslih, predhodno zdravljenih s kemoterapijo, ki je vključevala platino; lokalno napredovalega ali metastatskega urotelijskega raka pri odraslih, ki niso primerni za zdravljenje s kemoterapijo, ki vsebuje cisplatin in imajo tumorje z izraženostjo PD-L1 ≥ 10, ocenjeno s kombinirano pozitivno oceno (CPS); ponovljenega ali metastatskega ploščatoceličnega raka glave in vratu (HNSCC) pri odraslih, ki imajo tumorje z ≥ 50 % izraženostjo PD-L1 (TPS), in pri katerih je bolezen napredovala med zdravljenjem ali po zdravljenju s kemoterapijo, ki je vključevala platino in za prvo linijo zdravljenja metastatskega kolorektalnega raka z visoko mikrosatelitsko nestabilnostjo (MSI-H – microsatellite instability-high) ali s pomanjkljivim popravljanjem neujemanja pri podvojevanju DNA (dMMR - mismatch repair deficient) pri odraslih. Zdravilo KEYTRUDA je kot samostojno zdravljenje ali v kombinaciji s kemoterapijo s platino in 5-fluorouracilom (5-FU) indicirano za prvo linijo zdravljenja metastatskega ali neoperabilnega ponovljenega ploščatoceličnega raka glave in vratu pri odraslih, ki imajo tumorje z izraženostjo PD-L1 s CPS ≥ 1. Zdravilo KEYTRUDA je v kombinaciji s pemetreksedom in kemoterapijo na osnovi platine indicirano za prvo linijo zdravljenja metastatskega neploščatoceličnega NSCLC pri odraslih, pri katerih tumorji nimajo pozitivnih mutacij EGFR ali ALK; v kombinaciji s karboplatinom in bodisi paklitakselom bodisi nab-paklitakselom je indicirano za prvo linijo zdravljenja metastatskega ploščatoceličnega NSCLC pri odraslih; v kombinaciji z aksitinibom je indicirano za prvo linijo zdravljenja napredovalega raka ledvičnih celic (RCC) pri odraslih; v kombinaciji s kemoterapijo s platino in fluoropirimidinom indicirano za prvo linijo zdravljenja lokalno napredovalega neoperabilnega ali metastatskega raka požiralnika ali HER-2 negativnega adenokarcinoma gastroezofagealnega prehoda pri odraslih, ki imajo tumorje z izraženostjo PD-L1 s CPS ≥ 10. Odmerjanje in način uporabe: Testiranje PD-L1: Če je navedeno v indikaciji, je treba izbiro bolnika za zdravljenje z zdravilom KEYTRUDA na podlagi izraženosti PD-L1 tumorja potrditi z validirano preiskavo. Testiranje MSI-H/ dMMR pri bolnikih s CRC: Za samostojno zdravljenje z zdravilom KEYTRUDA je priporočljivo opraviti testiranje MSI-H/dMMR statusa tumorja z validirano preiskavo, da se izbere bolnike s CRC. Odmerjanje: Priporočeni odmerek zdravila KEYTRUDA pri odraslih je bodisi 200 mg na 3 tedne ali 400 mg na 6 tednov, apliciran z intravensko infuzijo v 30 minutah. Priporočeni odmerek zdravila KEYTRUDA za samostojno zdravljenje pri pediatričnih bolnikih s cHL, starih 3 leta ali več, je 2 mg/kg telesne mase (do največ 200 mg) na 3 tedne, apliciran z intravensko infuzijo v 30 minutah. Za uporabo v kombinaciji glejte povzetke glavnih značilnosti sočasno uporabljenih zdravil. Če se uporablja kot del kombiniranega zdravljenja skupaj z intravensko kemoterapijo, je treba zdravilo KEYTRUDA aplicirati prvo. Bolnike je treba zdraviti do napredovanja bolezni ali nesprejemljivih toksičnih učinkov. Pri adjuvantnem zdravljenju melanoma je treba zdravilo uporabljati do ponovitve bolezni, pojava nesprejemljivih toksičnih učinkov oziroma mora zdravljenje trajati do enega leta. Če je aksitinib uporabljen v kombinaciji s pembrolizumabom, se lahko razmisli o povečanju odmerka aksitiniba nad začetnih 5 mg v presledkih šest tednov ali več. Pri bolnikih starih ≥ 65 let, bolnikih z blago do zmerno okvaro ledvic, bolnikih z blago okvaro jeter prilagoditev odmerka ni potrebna. Odložitev odmerka ali ukinitev zdravljenja: Zmanjšanje odmerka zdravila KEYTRUDA ni priporočljivo. Za obvladovanje neželenih učinkov je treba uporabo zdravila KEYTRUDA zadržati ali ukiniti, prosimo, glejte celoten Povzetek glavnih značilnosti zdravila. Kontraindikacije: Preobčutljivost na učinkovino ali katero koli pomožno snov. Povzetek posebnih opozoril, previdnostnih ukrepov, interakcij in neželenih učinkov: Imunsko pogojeni neželeni učinki (pnevmonitis, kolitis, hepatitis, nefritis, endokrinopatije, neželeni učinki na kožo in drugi): Pri bolnikih, ki so prejemali pembrolizumab, so se pojavili imunsko pogojeni neželeni učinki, vključno s hudimi in smrtnimi primeri. Večina imunsko pogojenih neželenih učinkov, ki so se pojavili med zdravljenjem s pembrolizumabom, je bila reverzibilnih in so jih obvladali s prekinitvami uporabe pembrolizumaba, uporabo kortikosteroidov in/ali podporno oskrbo. Pojavijo se lahko tudi po zadnjem odmerku pembrolizumaba in hkrati prizadanejo več organskih sistemov. V primeru suma na imunsko pogojene neželene učinke je treba poskrbeti za ustrezno oceno za potrditev etiologije oziroma izključitev drugih vzrokov. Glede na izrazitost neželenega učinka je treba zadržati uporabo pembrolizumaba in uporabiti kortikosteroide – za natančna navodila, prosimo, glejte Povzetek glavnih značilnosti zdravila Keytruda. Zdravljenje s pembrolizumabom lahko poveča tveganje za zavrnitev pri prejemnikih presadkov čvrstih organov. Pri bolnikih, ki so prejemali pembrolizumab, so poročali o hudih z infuzijo povezanih reakcijah, vključno s preobčutljivostjo in anafilaksijo. Pembrolizumab se iz obtoka odstrani s katabolizmom, zato presnovnih medsebojnih delovanj zdravil ni pričakovati. Uporabi sistemskih kortikosteroidov ali imunosupresivov pred uvedbo pembrolizumaba se je treba izogibati, ker lahko vplivajo na farmakodinamično aktivnost in učinkovitost pembrolizumaba. Vendar pa je kortikosteroide ali druge imunosupresive mogoče uporabiti za zdravljenje imunsko pogojenih neželenih učinkov. Kortikosteroide je mogoče uporabiti tudi kot premedikacijo, če je pembrolizumab uporabljen v kombinaciji s kemoterapijo, kot antiemetično profilakso in/ali za ublažitev neželenih učinkov, povezanih s kemoterapijo. Ženske v rodni dobi morajo med zdravljenjem s pembrolizumabom in vsaj še 4 mesece po zadnjem odmerku pembrolizumaba uporabljati učinkovito kontracepcijo, med nosečnostjo in dojenjem se ga ne sme uporabljati. Varnost pembrolizumaba pri samostojnem zdravljenju so v kliničnih študijah ocenili pri 6.185 bolnikih z napredovalim melanomom, kirurško odstranjenim melanomom v stadiju III (adjuvantno zdravljenje), NSCLC, cHL, urotelijskim rakom, HNSCC ali CRC s štirimi odmerki (2 mg/kg telesne mase na 3 tedne, 200 mg na 3 tedne in 10 mg/ kg telesne mase na 2 ali 3 tedne). V tej populaciji bolnikov je mediani čas opazovanja znašal 7,6 mesece (v razponu od 1 dneva do 47 mesecev), najpogostejši neželeni učinki zdravljenja s pembrolizumabom so bili utrujenost (32 %), navzea (21 %) in diareja (21 %). Večina poročanih neželenih učinkov pri samostojnem zdravljenju je bila po izrazitosti 1. ali 2. stopnje. Najresnejši neželeni učinki so bili imunsko pogojeni neželeni učinki in hude z infuzijo povezane reakcije. Varnost pembrolizumaba pri kombiniranem zdravljenju s kemoterapijo so ocenili pri 1.437 bolnikih NSCLC, HNSCC ali rakom požiralnika, ki so v kliničnih študijah prejemali pembrolizumab v odmerkih 200 mg, 2 mg/kg telesne mase ali 10 mg/kg telesne mase na vsake 3 tedne. V tej populaciji bolnikov so bili najpogostejši neželeni učinki naslednji: navzea (55 %), anemija (51 %), utrujenost (39 %), zaprtost (37 %), zmanjšanje apetita (34 %), diareja (33 %), nevtropenija (29 %) in bruhanje (28 %). Pojavnost neželenih učinkov 3. do 5. stopnje je pri bolnikih z NSCLC pri kombiniranem zdravljenju s pembrolizumabom znašala 67 % in pri zdravljenju samo s kemoterapijo 66 %, pri bolnikih s HNSCC pri kombiniranem zdravljenju s pembrolizumabom 85 % in pri zdravljenju s kemoterapijo v kombinaciji s cetuksimabom 84 %, ter pri bolnikih z rakom požiralnika pri kombiniranem zdravljenju s pembrolizumabom 86 % in pri zdravljenju samo s kemoterapijo 83 %. Varnost pembrolizumaba v kombinaciji z aksitinibom so ocenili v klinični študiji pri 429 bolnikih z napredovalim rakom ledvičnih celic, ki so prejemali 200 mg pembrolizumaba na 3 tedne in 5 mg aksitiniba dvakrat na dan. V tej populaciji bolnikov so bili najpogostejši neželeni učinki diareja (54 %), hipertenzija (45 %), utrujenost (38 %), hipotiroidizem (35 %), zmanjšan apetit (30 %), sindrom palmarnoplantarne eritrodisestezije (28 %), navzea (28 %), zvišanje vrednosti ALT (27 %), zvišanje vrednosti AST (26 %), disfonija (25 %), kašelj (21 %) in zaprtost (21 %). Pojavnost neželenih učinkov 3. do 5. stopnje je bila med kombiniranim zdravljenjem s pembrolizumabom 76 % in pri zdravljenju s sunitinibom samim 71 %. Za celoten seznam neželenih učinkov, prosimo, glejte celoten Povzetek glavnih značilnosti zdravila. Način in režim izdaje zdravila: H – Predpisovanje in izdaja zdravila je le na recept, zdravilo se uporablja samo v bolnišnicah. Imetnik dovoljenja za promet z zdravilom: Merck Sharp & Dohme B.V. , Waarderweg 39, 2031 BN Haarlem, Nizozemska. Merck Sharp & Dohme inovativna zdravila d.o.o., Ameriška ulica 2, 1000 Ljubljana, tel: +386 1/ 520 42 01, fax: +386 1/ 520 43 50; Pripravljeno v Sloveniji, julij 2021; SI-KEY-00305 EXP: 07/2023 Samo za strokovno javnost. H - Predpisovanje in izdaja zdravila je le na recept, zdravilo pa se uporablja samo v bolnišnicah. Pred predpisovanjem, prosimo, preberite celoten Povzetek glavnih značilnosti zdravila Keytruda, ki je na voljo pri naših strokovnih sodelavcih ali na lokalnem sedežu družbe. CILJI ZDRAVLJENJA bolnic z napredovalim HR+/HER2- rakom dojk ESMO-MCBS † TOČK ASCO 2021 5 RAZLIKA JE! 1,2* NAJDALJŠE CELOKUPNO PREŽIVETJE in 3,4 IZBOLJŠANA ALI OHRANJENA KVALITETA ŽIVLJENJA KISQALI + zaviralec aromataze + goserelin, 1. red zdravljenja: mOS = 58,7 meseca * KISQALI + fulvestrant, 1. in 2. red zdravljenja: mOS = 53,7 meseca # KISQALI je edini zaviralec CDK4/6, ki dokazano podaljša življenje v različnih kombinacijah 1-6 (zaviralec aromataze ali fulvestrant) in redih zdravljenja ter hkrati izboljša ali ohranja kvaliteto življenja HR+ hormonsko odvisen rak dojk, HER2- rak dojk, negativen na receptorje humanega epidermalnega rastnega faktorja 2, Overall Survival, OS Celokupno preživetje, mediani OS, mOS mediana celokupnega preživetja. ESMO-MCBS, European Society for Medical Oncology Magnitude of Clinical Benefit Scale: ESMO lestvica obsega klinične koristi.† Zdravilo KISQALI je po ESMO-MCBS lestvici za študijo MONALEESA-7 prejelo največje možno število točk (5 točk)5. * MONALEESA-7 je bila randomizirana, dvojno slepa, s placebom nadzorovana multicentrična klinična študija faze III zdravljenja premenopavznih in perimenopavznih žensk z napredovalim HR+ HER2- rakom dojk, ki so poleg endokrinega zdravljenja prejemale še zdravilo Kisqali ali placebo. V raziskavo je bilo vključenih 672 bolnic, zdravilo Kisqali. je prejemalo 335 bolnic. V študiji je bil dosežen sekundarni cilj, dokazana je bila statistično značilna razlika med obema skupinama v dolžini preživetja bolnic1,6,7. Relativno znižanje tveganja za smrt je bilo 24% (razmerje ogroženosti = 0,76: 95% IZ [0,608; 0,956])1. V skupini bolnic, ki so prejemale kombinacijo zdravila KISQALI + zaviralec aromataze je bilo doseženo najdaljše mediano celokupno preživetje med vsemi raziskavami faze III v katere so bile vključene bolnice s HR+ HER2- napredovalim rakom dojk: 58,7 meseca1. # MONALEESA- 3 je bila randomizirana, dvojno slepa, s placebom nadzorovana multicentrična klinična študija faze III zdravljenja pomenopavznih žensk z napredovalim HR+ HER2- rakom dojk, ki so poleg fulvestranta prejemale še zdravilo Kisqali ali placebo v prvi ali drugi liniji zdravljenja. V študijo je bilo vključenih 726 bolnic, zdravilo Kisqali je prejemalo 484 bolnic. V študiji je bil dosežen sekundarni cilj, dokazana je bila statistično značilna razlika med obema skupinama v dolžini preživetja bolnic2,6,8. Relativno znižanje tveganja za smrt je bilo 28% (razmerje ogroženosti = 0,72: 95% IZ [0,568; 0,924])8. Podatki do presečnega datuma 30. oktober 2020 za skupino bolnic, ki so prejemale kombinacijo zdravila Kisqali in fulvestranta kažejo mediano celokupno preživetje, 53,7 meseca (razmerje ogroženosti 0,73: 95% IZ [0,59; 0,90]).2 Reference: 1. Tripathy D, Im S-A, Colleoni M, in sod. Updated overall survival (OS) results from the phase III MONALEESA-7 trial of pre- or perimenopausal patients with hormone receptor positive/human epidermal growth factor receptor 2 negative (HR+/HER2–) advanced breast cancer (ABC) treated with endocrine therapy (ET) +/- ribociclib. Predstavljeno na: San Antonio Breast Cancer Symposium; 8-12. december 2020, 2020; San Antonio, Texas. Poster PD2-04. 2. Slamon D, Neven P, Chia S, in sod. Updated overall survival (OS) results from the phase III MONALEESA-3 trial of postmenopausal patients (pts) with HR+/HER2- advanced breast cancer (ABC) treated with fulvestrant ± placebo. Predstavljeno na kongresu American Society of Clinical Oncology (ASCO) 2021, 4-8. junij 2021, virtualni kongres. 3. Harbeck N, Vazquez RV, Franke F, in sod. Ribociclib+ tamoxifen or a non-steroidal aromatase inhibitor in premenopausal patients with hormone receptor-positive, HER2-negative advanced breast cancer: MONALEESA-7 patient-reported outcomes. Predstavljeno na: European Society for Medical Oncology Congress; 19- 23 oktober 2018; Munchen, Nemčija. 4. Fasching PA, Esteva FJ, Pivot X, in sod. Patient-reported outcomes in advanced breast cancer treated with ribociclib + fulvestrant: results from MONALEESA-3. Predstavljeno na: European Society for Medical Oncology Congress; 19-23. oktober, Munchen, Nemčija. 5. Cardoso F, Paluch-Shimon S, Senkus E, in sod. 5th ESOESMO international consensus guidelines for advanced breast cancer (ABC5) 2020; 31 (12): 1623-1649. 6. Povzetek glavnih značilnosti zdravila Kisqali. Oktober 2020. 7. Im SA, Lu YS, Bardia A, in sod. Overall Survival with Ribociclib plus Endocrine Therapy in Breast Cancer. N Engl J Med 2019; 381:307-16. 8. Slamon DJ, Neven P, Chia S, in sod. Overall Survival with Ribociclib plus Fulvestrant in Advanced Breast Cancer. N Engl J Med 2020; 382:514-524. SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA KISQALI® Za to zdravilo se izvaja dodatno spremljanje varnosti. Zdravstvene delavce naprošamo, da poročajo o katerem koli domnevnem neželenem učinku zdravila. Glejte poglavje 4.8 povzetka glavnih značilnosti zdravila, kako poročati o neželenih učinkih. Ime zdravila: Kisqali 200 mg filmsko obložene tablete. Sestava: Ena tableta vsebuje ribociklibijev sukcinat v količini, ki ustreza 200 mg ribocikliba. Indikacija: Zdravilo Kisqali je v kombinaciji z zaviralcem aromataze ali fulvestrantom indicirano za zdravljenje žensk z lokalno napredovalim ali metastatskim rakom dojk, ki je hormonsko odvisen (HR pozitiven) in negativen na receptorje humanega epidermalnega rastnega faktorja 2 (HER2 negativen), in sicer kot začetno hormonsko zdravljenje ali pri ženskah, ki so predhodno že prejemale hormonsko zdravljenje. Pri ženskah pred menopavzo ali v perimenopavzi je treba hormonsko zdravljenje uporabljati skupaj z agonistom gonadoliberina (LHRH-luteinizing hormone releasing hormone). Odmerjanje in način uporabe: Zdravljenje mora uvesti zdravnik, ki ima izkušnje z uporabo zdravil proti raku. Priporočeni odmerek je 600 mg (tri 200‑miligramske tablete) ribocikliba 1x/dan 21 dni zaporedoma, čemur sledi 7 dni brez zdravljenja, tako da celotni ciklus traja 28 dni. Zdravljenje je treba nadaljevati, dokler ima bolnik od zdravljenja klinično korist oz. do pojava nesprejemljivih toksičnih učinkov. Kisqali je treba uporabljati skupaj z 2,5 mg letrozola ali drugim zaviralcem aromataze ali s 500 mg fulvestranta. Zaviralec aromataze je treba jemati peroralno 1x/dan neprekinjeno vseh 28 dni ciklusa. Fulvestrant je treba odmerjati intramuskularno 1., 15. in 29. dan ciklusa, nato pa 1x/mesec. Za več podatkov glejte povzetek glavnih značilnosti zdravila za zaviralec aromataze oz. fulvestrant. Ženske pred menopavzo ali v perimenopavzi morajo prejemati tudi katerega od agonistov gonadoliberina v skladu z lokalno klinično prakso. Kisqali je treba jemati peroralno 1x/dan skupaj s hrano ali brez nje. Bolniki naj vzamejo odmerek zdravila vsak dan ob približno istem času, najbolje zjutraj. Tablete je treba pogoltniti cele in se jih pred zaužitjem ne sme gristi, drobiti ali lomiti. Tablet, ki so razlomljene, zdrobljene ali kako drugače poškodovane, se ne sme zaužiti. Če bolnik po zaužitju odmerka bruha ali pozabi vzeti odmerek, na ta dan ne sme vzeti dodatnega odmerka. Naslednji predpisani odmerek mora vzeti ob običajnem času. Prilagajanje odmerkov: Obvladovanje hudih ali nesprejemljivih neželenih dogodkov zdravila lahko vključuje prekinitev jemanja zdravila, znižanje odmerka ali ukinitev zdravila Kisqali. Ob prvem zmanjšanju odmerek zmanjšamo na 400 mg/dan (dve 200‑miligramski tableti), ob drugem zmanjšanju pa na 200 mg/dan (ena 200‑miligramska tableta). Če bi bilo treba odmerek zmanjšati na manj kot 200 mg/dan, je treba zdravljenje ukiniti. Za priporočila glede prekinitve jemanja zdravila, zmanjšanje odmerka ali ukinitev zdravljenja v primerih, ko je to potrebno za obvladovanje določenih neželenih dogodkov zdravila, prosimo glejte povzetek glavnih značilnosti zdravila. Za prilagajanje odmerkov in druge pomembne podatke v primeru toksičnega delovanja glejte povzetek glavnih značilnosti zdravila za sočasno uporabljeni zaviralec aromataze, fulvestrant oz. agonist gonadoliberina. Okvara ledvic: Pri bolnikih z blago ali zmerno okvaro ledvic prilagajanje odmerka ni potrebno. Pri bolnikih s hudo okvaro ledvic (aGFR 15 do < 30 ml/min) je priporočen začetni odmerek 200 mg. Okvara jeter: Bolnikom z blago okvaro jeter (Child‑Pugh razreda A) odmerka ni potrebno prilagajati. Pri bolnikih z zmerno (Child‑Pugh razreda B) ali hudo okvaro jeter (Child‑Pugh razreda C) je priporočeni začetni odmerek zdravila Kisqali 400 mg 1x/dan. Pediatrična populacija: Varnost in učinkovitost zdravila pri otrocih in mladostnikih, starih manj kot 18 let, nista bili dokazani. Starostniki: Pri bolnikih, ki so stari več kot 65 let, prilagajanje odmerka ni potrebno. Kontraindikacije: Preobčutljivost na učinkovino, arašide, sojo ali katero koli pomožno snov. Posebna opozorila in previdnostni ukrepi: Kritična visceralna bolezen: Učinkovitosti in varnosti ribocikliba pri bolnikih s kritično visceralno boleznijo niso proučevali. Nevtropenija in hepatobiliarna toksičnost: Pregled celotne krvne slike in vrednosti jetrnih testov je treba opraviti pred začetkom zdravljenja, nato v prvih 2 ciklusih vsaka 2 tedna, v naslednjih 4 ciklusih na začetku vsakega ciklusa, nato pa kot je klinično indicirano. Če pride do nenormalnih vrednosti jetrnih testov stopnje ≥ 2, so priporočene pogostejše meritve jetrnih testov. Za bolnike z zvišanjem vrednosti AST/ALT stopnje ≥3 ob izhodišču priporočila za odmerjanje niso dognana. Glede na to, kako močno je izražena nevtropenija ali zvišane vrednosti aminotransferaz, je morda treba odmerjanje zdravila Kisqali prekiniti, zmanjšati odmerek ali zdravljenje ukiniti. Podaljšanje intervala QT: Pred začetkom zdravljenja je treba posneti EKG. Zdravljenje je mogoče začeti samo pri bolnikih s trajanjem intervala QTcF manj kot 450 ms. EKG je treba ponovno posneti približno 14. dan prvega ciklusa in na začetku drugega ciklusa, nato pa kot je klinično indicirano. V primeru, da v času zdravljenja pride do podaljšanja intervala QTcF, je priporočeno pogostejše snemanje EKG. Ustrezno spremljanje koncentracij elektrolitov v serumu (vključno s koncentracijami kalija, kalcija, fosforja in magnezija) je treba izvajati pred začetkom zdravljenja, nato na začetku prvih 6 ciklusov in kasneje kot je klinično indicirano. Kakršnekoli nepravilnosti je treba odpraviti pred začetkom oz. med potekom zdravljenja z zdravilom Kisqali. Uporabi zdravila Kisqali se je treba izogibati pri bolnikih s prisotnim podaljšanjem intervala QTc ali s povečanim tveganjem za podaljšanje intervala QTc. To vključuje bolnike s sindromom podaljšanega intervala QT, z neurejenim ali pomembnim srčnim obolenjem, kar vključuje nedaven miokardni infarkt, kongestivno popuščanje srca, nestabilno angino pektoris in bradiaritmije ter bolnike z elektrolitskimi nepravilnostmi. Izogibati se je treba sočasni uporabi zdravila Kisqali z zdravili, za katera je znano, da lahko podaljšajo interval QT, kot so antiaritmiki (med drugim amiodaron, dizopiramid, prokainamid, kinidin in sotalol) ter druga zdravila, za katera je znano, da podaljšujejo interval QT (med drugim klorokin, halofantrin, klaritromicin, ciprofloksacin, levofloksacin, azitromicin, haloperidol, metadon, moksifloksacin, bepridil, pimozid in intravenski ondansetron). Zdravila Kisqali prav tako ni priporočeno uporabljati v kombinaciji s tamoksifenom. Če se zdravljenju z močnim zaviralcem CYP3A4 ni mogoče izogniti, je treba odmerek zdravila Kisqali zmanjšati na 400 mg 1x/dan. Glede na izmerjeno podaljšanje intervala QT v času zdravljenja je morda treba odmerjanje zdravila Kisqali prekiniti, zmanjšati odmerek ali zdravljenje ukiniti. Hude kožne reakcije: Poročali so o pojavu toksične epidermalne nekrolize (TEN). Če se pojavijo znaki in simptomi, ki lahko pomenijo, da gre za hudo kožno reakcijo (na primer progresiven generaliziran kožni izpuščaj, pogosto z mehurji, ali lezijami sluznice), je treba zdravljenje takoj prekiniti. Intersticijska pljučna bolezen/pnevmonitis: Glede na izraženost intersticijske pljučne bolezni/pnevmonitisa, ki se lahko končata tudi s smrtjo bolnika, bo v skladu s priporočilom v povzetku glavnih značilnosti zdravila morda potrebno odmerjanje zdravila Kisqali prekiniti, zmanjšati odmerek ali zdravljenje ukiniti. Bolnike je treba spremljati glede pljučnih simptomov, ki bi lahko nakazovali na intersticijsko pljučno bolezen/pnevmonitis, in lahko vključujejo hipoksijo, kašelj in dispnejo. Zvišanje kreatinina: Če v času zdravljenja pride do zvišanja vrednosti kreatinina v krvi, je priporočeno izvesti dodatno oceno ledvične funkcije za izključitev okvare ledvic. Okvara ledvic: Pri začetnem odmerku 200 mg pri bolnikih s hudo okvaro ledvic so ocenili, da je izpostavljenost za približno 45 % nižja kot pri standardnem začetnem odmerku pri bolnikih z normalnim delovanjem ledvic. Učinkovitosti pri tem začetnem odmerku niso preučevali. Pri bolnikih s hudo okvaro ledvic je potrebna previdnost s skrbnim spremljanjem glede znakov toksičnega delovanja. Sojin lecitin: Zdravilo vsebuje sojin lecitin. Bolniki s preobčutljivostjo na arašide ali sojo ne smejo jemati zdravila Kisqali. Plodnost, nosečnost in dojenje: Pred začetkom zdravljenja je treba preveriti status nosečnosti. Ženskam v rodni dobi je treba svetovati, naj v času zdravljenja z zdravilom Kisqali in še najmanj 21 dni po prejemu zadnjega odmerka uporabljajo učinkovito kontracepcijsko metodo. Bolnice, ki jemljejo zdravilo Kisqali, ne smejo dojiti še najmanj 21 dni po prejemu zadnjega odmerka. Glede na ugotovitve študij pri živalih lahko zdravilo Kisqali zmanjša plodnost pri reproduktivno sposobnih moških. Vpliv na sposobnost vožnje in upravljanja strojev: Zdravilo ima lahko blag vpliv na sposobnost vožnje in upravljanja strojev. Bolnike je treba opozoriti, naj bodo pri vožnji in upravljanju strojev previdni, če imajo v času zdravljenja težave z utrujenostjo, omotičnostjo ali vrtoglavico. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij: Snovi, ki lahko zvišajo koncentracijo ribocikliba v plazmi: Izogibati se je treba sočasni uporabi močnih zaviralcev CYP3A4, med drugim klaritromicinu, indinavirju, itrakonazolu, ketokonazolu, lopinavirju, ritonavirju, nefazodonu, nelfinavirju, posakonazolu, sakvinavirju, telaprevirju, telitromicinu, verapamilu in vorikonazolu. Za sočasno uporabo je treba razmisliti o izbiri drugih zdravil z manjšim potencialom za zaviranje CYP3A4, bolnike pa je treba spremljati glede neželenih dogodkov v povezavi z ribociklibom. Če mora bolnik sočasno z ribociklibom prejemati močan zaviralec CYP3A4, je treba odmerek zdravila Kisqali zmanjšati na 400 mg 1x/dan. Pri bolnikih, pri katerih je odmerek že zmanjšan na 400 mg/dan, je treba odmerek zmanjšati na 200 mg, pri bolnikih, pri katerih je odmerek ribocikliba že zmanjšan na 200 mg dnevno, pa je treba zdravljenje z zdravilom Kisqali prekiniti. Zaradi interindividualne variabilnosti priporočeno prilagajanje odmerjanja morda ni najboljše za vse bolnike, zato je priporočeno skrbno spremljanje bolnikov glede znakov toksičnega delovanja. Če bolnik preneha jemati zdravilo, ki je močan zaviralec, je treba odmerek zdravila Kisqali prilagoditi in po najmanj 5 razpolovnih dobah močnega zaviralca CYP3A4 spet začeti z odmerkom zdravila Kisqali, ki ga je bolnik prejemal pred začetkom uporabe močnega zaviralca CYP3A4. Po začetku sočasne uporabe šibkih ali zmernih zaviralcev CYP3A4 odmerka ribocikliba ni treba prilagajati, priporočeno pa je spremljanje bolnikov glede neželenih dogodkov v povezavi z ribociklibom. Bolnikom je treba naročiti, naj se izogibajo uživanju grenivk in njihovega soka, ker lahko povečajo izpostavljenost ribociklibu. Snovi, ki lahko znižajo koncentracijo ribocikliba v plazmi: Sočasna uporaba močnih induktorjev CYP3A4 lahko zmanjša izpostavljenost zdravilu, to pa lahko predstavlja tveganje za manjšo učinkovitost zdravila. Izogibati se je treba sočasni uporabi močnih induktorjev CYP3A4, med drugim fenitoina, rifampicina, karbamazepina in šentjanževke (Hypericum perforatum). Za sočasno uporabo je treba razmisliti o izbiri drugega zdravila, ki ne inducira oziroma ima manjši potencial za indukcijo CYP3A4. Učinka zmernega induktorja CYP3A4 na izpostavljenost ribociklibu niso proučevali. Sočasna uporaba zmernega induktorja CYP3A4 lahko privede do zmanjšane izpostavljenosti ribociklibu, to pa lahko predstavlja tveganje za manjšo učinkovitost, še posebej pri bolnikih, ki se zdravijo z ribociklibom v odmerku 400 mg ali 200 mg 1x/dan. Snovi, na katerih koncentracijo v plazmi lahko vpliva zdravilo Kisqali: Ribociklib je zmeren do močan zaviralec CYP3A4, zato lahko pride do interakcij z zdravili, ki so substrati oz. jih presnavlja CYP3A4, kar lahko povzroči zvišanje koncentracij sočasno uporabljenih zdravil v serumu. Pri sočasnem odmerjanju ribocikliba z drugimi zdravili je praviloma treba pregledati povzetek glavnih značilnosti drugega zdravila in poiskati priporočila za sočasno odmerjanje z zaviralci CYP3A4. Pri sočasni uporabi z občutljivimi substrati CYP3A4, ki imajo nizek terapevtski indeks, je priporočena previdnost. Pri teh substratih, med drugim pri alfentanilu, ciklosporinu, everolimusu, fentanilu, sirolimusu in takrolimusu, je v nekaterih primerih treba zmanjšati njihov odmerek, saj ribociklib lahko poveča izpostavljenost tem snovem. Izogibati se je treba sočasni uporabi ribocikliba v odmerku 600 mg skupaj z naslednjimi substrati CYP3A4: alfuzosin, amiodaron, cisaprid, pimozid, kinidin, ergotamin, dihidroergotamin, kvetiapin, lovastatin, simvastatin, sildenafil, midazolam in triazolam. Pri uporabi klinično ustreznega odmerka 600 mg je mogoče pričakovati le šibak zaviralni učinek ribocikliba na substrate CYP1A2 (< 2‑kratno povečanje AUC). Snovi, ki so substrati prenašalcev: Vrednotenje podatkov in vitro raziskav kaže, da ima ribociklib potencial za zaviranje aktivnosti prenašalcev P-gp, BCRP, OATP1B1/1B3, OCT1, OCT2, MATE1 in BSEP. Pri sočasnem zdravljenju s snovmi, ki so občutljivi substrati teh prenašalcev in imajo nizek terapevtski indeks, med drugim z digoksinom, pitavastatinom, pravastatinom, rosuvastatinom in metforminom, je priporočena previdnost in spremljanje bolnikov glede toksičnega delovanja. Zdravila, ki zvišujejo pH v želodcu: Pri populacijski farmakokinetični analizi in nekompartmentalni farmakokinetični analizi pri sočasni uporabi niso opažali sprememb v absorpciji ribocikliba. Medsebojno delovanje med ribociklibom in letrozolom oz. anastrozolom oz. fulvestrantom oz. tamoksifenom: Med ribociklibom in letrozolom oz. med ribociklibom in anastrozolom ne prihaja do medsebojnega delovanja pri sočasnem odmerjanju obeh zdravil. Po podatkih iz klinične študije fulvestrant nima klinično pomembnega vpliva na izpostavljenost ribociklibu pri sočasnem odmerjanju, po sočasnem odmerjanju ribocikliba in tamoksifena pa se je izpostavljenost tamoksifenu povečala na približno 2‑kratno vrednost. Neželeni učinki: Zelo pogosti (≥ 1/10): okužbe, nevtropenija, levkopenija, anemija, limfopenija, zmanjšan apetit, glavobol, omotičnost, dispneja, kašelj, navzea, diareja, bruhanje, obstipacija, stomatitis, bolečine v trebuhu, dispepsija, alopecija, izpuščaj, pruritus, bolečine v hrbtu, utrujenost, periferni edemi, astenija, zvišana telesna temperatura, nenormalne vrednosti jetrnih testov. Pogosti (≥ 1/100 do < 1/10): trombocitopenija, febrilna nevtropenija, hipokalciemija, hipokaliemija, hipofosfatemija, vrtoglavica, močnejše solzenje, suhe oči, sinkopa, motnje okušanja, hepatotoksičnost, eritem, suha koža, vitiligo, suha usta, orofaringealna bolečina, zvišana vrednost kreatinina v krvi, podaljšan interval QT v elektrokardiogramu. Neznana pogostnost: toksična epidermalna nekroliza (TEN). Imetnik dovoljenja za promet z zdravilom: Novartis Europharm Limited, Vista Building, Elm Park, Merrion Road, Dublin 4, Irska. Dodatne informacije in literatura: Novartis Pharma Services Inc., Podružnica v Sloveniji, Verovškova ulica 57, 1000 Ljubljana. Način/režim izdajanja zdravila: Rp/Spec. Pred predpisovanjem natančno preberite zadnji odobreni povzetek glavnih značilnosti zdravila. Datum zadnje revizije skrajšanega povzetka glavnih značilnosti zdravila: november 2020. Novartis Pharma Services Inc., Podružnica v Sloveniji Verovškova ulica 57, 1000 Ljubljana, tel.: 01 300 75 50 Datum priprave materiala: junij 2021 Samo za strokovno javnost. SI-2021-KIS-099 Prva terapija za zdravljenje odraslih bolnikov z metastatskim ali lokalno napredovalim ploščatoceličnim karcinomom kože (PCKK), ki niso kandidati za kurativni kirurški poseg ali kurativno obsevanje. 1,2 Zaviralec PD-1: spodbuja bolnikov imunski p rotitumorski odziv za izboljšanje rezultatov zdravljenja 3 PD-1, receptor programirane celične smrti 1 Pred predpisovanjem prosimo preberite celoten povzetek glavnih značilnosti zdravila. SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA ▼Za to zdravilo se izvaja dodatno spremljanje varnosti. Tako bodo hitreje na voljo nove informacije o njegovi varnosti. Zdravstvene delavce naprošamo, da poročajo o katerem koli domnevnem neželenem učinku zdravila. Ime zdravila: LIBTAYO 350 mg koncentrat za raztopino za infundiranje. Sestava: En mililiter koncentrata vsebuje 50 mg cemiplimaba. Ena viala vsebuje 350 mg cemiplimaba v 7 ml raztopine. Terapevtske indikacije: Ploščatocelični karcinom kože: Zdravilo LIBTAYO je kot samostojno zdravljenje (monoterapija) indicirano za zdravljenje odraslih bolnikov z metastatskim ali lokalno napredovalim ploščatoceličnim karcinomom kože (mPCKK ali lnPCKK), ki niso kandidati za kurativni kirurški poseg ali kurativno obsevanje. Bazalnocelični karcinom: Zdravilo LIBTAYO je kot samostojno zdravljenje (monoterapija) indicirano za zdravljenje odraslih bolnikov z lokalno napredovalim ali metastatskim bazalnoceličnim karcinomom (1aBCK ali mBCK), pri katerih je bolezen napredovala kljub uporabi zaviralca signalne poti Hedgehog (HHI) ali ga ne prenašajo. Nedrobnocelični pljučni rak: Zdravilo LIBTAYO je kot samostojno zdravljenje (monoterapija) indicirano za prvo linijo zdravljenja odraslih bolnikov z nedrobnoceličnim pljučnim rakom (NSCLC – Non-Small Cell Lung Cancer), katerih tumorji izražajo PD-L1 (v ≥ 50 % tumorskih celic) brez aberacij EGFR, ALK ali ROSI z: lokalno napredovalim NSCLC, ki niso primerni za definitivno kemoradiacijo, ali metastatskim NSCLC. Odmerjanje in način uporabe: Zdravljenje mora uvesti in nadzorovati zdravnik, izkušen na področju zdravljenja raka. Testiranje PD-L1 pri bolnikih z NSCLC: Za zdravljenje s cemiplimabom kot monoterapijo je treba bolnike izbrati na podlagi validiranega testa izražanja PD-L1 v tumorju. Priporočeni odmerek: Priporočeni odmerek cemiplimaba je 350 mg na 3 tedne v 30 minutni intravenski infuziji. Zdravljenje se sme nadaljevati do napredovanja bolezni ali nesprejemljivih toksičnih učinkov. Prilagoditve odmerka: Zmanjšanja odmerka niso priporočena. Glede na varnost in prenašanje pri posameznem bolniku je lahko potrebna odložitev odmerka ali prenehanje uporabe. Za priporočene prilagoditve za obvladovanje neželenih učinkov glejte celoten Povzetek glavnih značilnosti zdravila. Posebne populacije: Pediatrična populacija: Varnost in učinkovitost zdravila LIBTAYO pri otrocih in mladostnikih, mlajših od 18 let, nista ugotovljeni. Starejše osebe, okvara ledvic, okvara jeter: odmerka ni treba prilagoditi. Način uporabe: Zdravilo LIBTAYO je namenjeno intravenski uporabi. Daje se v intravenski infuziji v obdobju 30 minut po intravenski liniji, ki vsebuje sterilen, nepirogen filter (v sami liniji ali kot dodatek), ki malo veže beljakovine (velikost por od 0,2 do 5 mikronov). Po isti infuzijski liniji se ne sme istočasno dajati drugih zdravil. Kontraindikacije: Preobčutljivost na učinkovino ali katero koli pomožno snov. Posebna opozorila in previdnostni ukrepi: Sledljivost: Z namenom izboljšanja sledljivosti bioloških zdravil je treba jasno zabeležiti ime in številko serije uporabljenega zdravila. Imunsko pogojeni neželeni učinki: Med uporabo cemiplimaba so opažali hude imunsko pogojene neželene učinke, tudi s smrtnim izidom. Pri bolnikih, zdravljenih s cemiplimabom ali drugimi zaviralci PD-1/PD-L1, se lahko sočasno pojavijo imunski neželeni učinki, ki vplivajo na več telesnih sistemov, na primer miozitis in miokarditis ali miastenija gravis. Za obvladanje imunsko pogojenih neželenih učinkov je treba prilagoditi odmerek cemiplimaba, nadomestno hormonsko zdravljenje (če je klinično indicirano) in kortkosteroide. Odvisno od izrazitosti neželenega učinka je treba uporabo cemiplimaba začasno prekiniti ali za stalno prenehati. Imunsko pogojeni pnevmonitis: Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojeni pnevmonitis, opredeljen s potrebo po uporabi kortikosteroidov in brez jasne alternativne etiologije, vključno s primeri s smrtnim izidom. Imunsko pogojeni kolitis: Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojeno drisko ali kolitis, opredeljena s potrebo po uporabi kortikosteroidov in brez jasne alternativne etiologije. Imunsko pogojeni hepatitis: Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojeni hepatitis, opredeljen s potrebo po uporabi kortikosteroidov in brez jasne alternativne etiologije, vključno s primeri s smrtnim izidom. Imunsko pogojene endokrinopatije: Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojene endokrinopatije, opredeljene kot med zdravljenjem nastale endokrinopatije brez jasne alternativne etiologije. Motnje v delovanju ščitnice (hipotiroidizem/hipertiroidizem/tiroiditis): Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojene motnje v delovanju ščitnice. Tiroiditis se lahko pojavi s spremembo testov delovanja ščitnice ali brez nje. Hipertiroidizmu lahko sledi hipotiroidizem. Hipofizitis: Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojeni hipofizitis. Nadledvična insuficienca: Pri bolnikih, ki so prejemali cemiplimab, so opažali nadledvično insuficienco. Sladkorna bolezen tipa 1: Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojeno sladkorno bolezen tipa 1, vključno z diabetično ketoacidozo. Imunsko pogojeni neželeni učinki na kožo: Med zdravljenjem s cemiplimabom so poročali o imunsko pogojenih neželenih učinkih na kožo, opredeljenih s potrebo po uporabi sistemskih kortikosteroidov in brez jasne alternativne etiologije; med njimi so bili hudi neželeni učinki na kožo, na primer Stevens-Johnsonov sindrom (SJS) in toksična epidermalna nekroliza (TEN) (v nekaterih primerih s smrtnim izidom), in druge kožne reakcije, na primer izpuščaj, multiformni eritem in pemfigoid. Imunsko pogojeni nefritis: Pri bolnikih, ki so prejemali cemiplimab, so opažali imunsko pogojeni nefritis, opredeljen s potrebo po uporabi kortikosteroidov in brez jasne alternativne etiologije. Drugi imunsko pogojeni neželeni učinki: Pri bolnikih, ki so prejemali cemiplimab, so opažali še druge življenjsko nevarne in smrtne imunsko pogojene neželene učinke, med njimi paraneoplastični encefalomielitis, meningitis in miozitis. Zdravljenje s cemiplimabom lahko pri prejemnikih presadkov parenhimskih organov poveča tveganje za zavrnitev. V obdobju po prihodu na trg so pri bolnikih, ki so prejemali druge zaviralce PD-1/PD-L1 obenem z alogensko presaditvijo hematopoetskih matičnih celic, poročali o primerih bolezni presadka proti gostitelju. Z infundiranjem povezane reakcije: Cemiplimab lahko povzroči resne ali življenjsko nevarne z infundiranjem povezane reakcije. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij: Uporabi sistemskih kortikosteroidov ali imunosupresivov pred uvedbo cemiplimaba se je treba izogibati, razen fizioloških odmerkov sistemskih kortikosteroidov (≤ 10 mg/dan prednizolona ali enakovredno), ker lahko motijo farmakodinamično aktivnost in učinkovitost cemiplimaba. Vendar pa je kortikosteroide ali druge imunosupresive mogoče uporabiti po začetku zdravljenja s cemiplimabom za zdravljenje imunsko pogojenih neželenih učinkov. Plodnost, nosečnost in dojenje: Ženske v rodni dobi morajo med zdravljenjem s cemiplimabom in vsaj še 4 mesece po zadnjem odmerku cemiplimaba uporabljati učinkovito kontracepcijo. Cemiplimab ni priporočljiv med nosečnostjo in za ženske v rodni dobi, ki ne uporabljajo učinkovite kontracepcije, razen če klinična korist odtehta možno tveganje. Če se ženska odloči za zdravljenje s cemiplimabom, ji je treba svetovati, da med zdravljenjem s cemiplimabom in vsaj še 4 mesece po zadnjem odmerku ne sme dojiti. Vpliv na sposobnost vožnje in upravljanja strojev: Po zdravljenju s cemiplimabom so poročali o utrujenosti. Neželeni učinki: Zelo pogosti: okužba zgornjih dihal, anemija, zmanjšan apetit, kašelj, slabost, driska, zaprtje, izpuščaj, pruritus, mišično-skeletna bolečina, utrujenost. Pogosti: okužba sečil, z infundiranjem povezane reakcije, hipotiroidizem, hipertiroidizem, glavobol, periferna nevropatija, hipertenzija, dispneja, pnevmonitis, bolečina v trebuhu, bruhanje, stomatitis, kolitis, hepatitis, artritis, nefritis, zvišana aspartat-aminotransferaza, zvišana alanin-aminotransferaza, zvišana alkalna fosfataza v krvi, zvišan kreatinin v krvi. Občasni: sjögrenov sindrom, imunsko pogojena trombocitopenična purpura, nadledvična insuficienca, tiroiditis, sladkorna bolezen tipa 1, hipofizitis, meningitis, encefalitis, miastenija gravis, praneoplastični encefalomielitis, kronična vnetna demielinizirajoča poliradikulonevropatija, keratitis, miokarditis, perikarditis, šibkost mišic, miozitis, revmatska polimialgija, zvišan ščitnični hormon v krvi, zvišane transaminase, zvišan bilirubin v krvi, znižan ščitnični hormon v krvi. Preveliko odmerjanje: V primeru prevelikega odmerjanja naj se bolnike natančno kontrolira glede znakov in simptomov neželenih učinkov in uvede ustrezno simptomatsko zdravljenje. Način in režim izdaje zdravila: H-Predpisovanje in izdaja zdravila je le na recept, zdravilo pa se uporablja samo v bolnišnicah. Imetnik dovoljenja za promet z zdravilom: Regeneron Ireland Designated Activity Company (DAC), One Warrington Place, Dublin 2, D02 HH27, Irska. Datum zadnje revizije besedila: 21.06.2021 SAMO ZA STROKOVNO JAVNOST MAT-SI-2000079-5.0-09/2021 1. Libtayo (cemiplimab) Povzetek glavnih značilnosti zdravila, www.ema.europa.com, datum zadnjega podaljšanja 08.03.2021 2. www.nice.org.uk, technology appraisal guidance TA592. dostop 07.08.2019. 3. www.cancer.gov/publications/dictionaries/cancer-terms/def/pd-1, dostop 07.08.2019 ZAUPANJE, ZGRAJENO NA MOČI Za zdravljenje lokalno napredovalega ali metastatskega HR+/HER2- raka dojk: • v kombinaciji z zaviralcem aromataze, • v kombinaciji s fulvestrantom pri ženskah, ki so prejele predhodno endokrino zdravljenje. Pri ženskah v pred- in perimenopavzi je treba endokrino zdravljenje kombinirati z agonistom gonadoliberina (LHRH – Luteinizing Hormone–Releasing Hormone). BISTVENI PODATKI IZ POVZETKA GLAVNIH ZNAČILNOSTI ZDRAVILA IIBRANCE 75 mg, 100 mg, 125 mg trde kapsule(1) IBRANCE 75 mg, 100 mg, 125 mg filmsko obložene tablete(2) Sestava in oblika zdravila: (1) Ena trda kapsula vsebuje 75 mg, 100 mg ali 125 mg palbocikliba in 56 mg, 74 mg ali 93 mg laktoze (v obliki monohidrata). (2) Ena filmsko obložena tableta vsebuje 75 mg, 100 mg ali 125 mg palbocikliba. Indikacije: Zdravljenje lokalno napredovalega ali metastatskega na hormonske receptorje (HR – Hormone Receptors) pozitivnega in na receptorje humanega epidermalnega rastnega faktorja 2 (HER2 – Human Epidermal growth factor Receptor 2) negativnega raka dojk: v kombinaciji z zaviralcem aromataze ali v kombinaciji s fulvestrantom pri ženskah, ki so prejele predhodno endokrino zdravljenje. Pri ženskah v pred- in perimenopavzi je treba endokrino zdravljenje kombinirati z agonistom gonadoliberina. Odmerjanje in način uporabe: Zdravljenje mora uvesti in nadzorovati zdravnik, ki ima izkušnje z uporabo zdravil za zdravljenje rakavih bolezni. Priporočeni odmerek je 125 mg enkrat na dan 21 zaporednih dni, sledi 7 dni brez zdravljenja (shema 3/1), celotni cikel traja 28 dni. Zdravljenje je treba nadaljevati, dokler ima bolnik od zdravljenja klinično korist ali dokler se ne pojavi nesprejemljiva toksičnost. Pri sočasnem dajanju s palbociklibom je treba zaviralec aromataze dajati v skladu s shemo odmerjanja, ki je navedena v Povzetku glavnih značilnosti zdravila (PGZZ). Pri sočasnem dajanju s palbociklibom je priporočeni odmerek fulvestranta 500 mg intramuskularno 1., 15. in 29. dan ter nato enkrat na mesec, glejte PGZZ za fulvestrant. Prilagajanja odmerkov: Za prilagajanja odmerkov zaradi hematološke toksičnosti glejte preglednico 2, zaradi nehematološke toksičnosti pa preglednico 3 v PGZZ‑ju. Pri bolnikih s hudo intersticijsko boleznijo pljuč (ILD)/pnevmonitisom je treba zdravljenje trajno prekiniti. Posebne skupine bolnikov: Starejši: Prilagajanje odmerka ni potrebno. Okvara jeter ali ledvic: Pri bolnikih z blago ali zmerno okvaro jeter ali blago, zmerno ali hudo okvaro ledvic prilagajanje odmerka ni potrebno. Pri bolnikih s hudo okvaro jeter je priporočeni odmerek 75 mg enkrat na dan po shemi 3/1. Pediatrična populacija: Varnost in učinkovitost pri otrocih in mladostnikih, starih < 18 let, nista bili dokazani. Način uporabe: Peroralna uporaba. (1) Jemanje s hrano, priporočljivo z obrokom. (2) Tablete se lahko jemlje s hrano ali brez nje. (1, 2) Ne smemo jemati z grenivko ali grenivkinim sokom. Kapsule oz. tablete zdravila je treba pogoltniti cele. Kontraindikacije: Preobčutljivost na učinkovino ali katerokoli pomožno snov. Uporaba pripravkov s šentjanževko. Posebna opozorila in previdnostni ukrepi: Ženske v pred- in perimenopavzi: Kadar zdravilo uporabljamo v kombinaciji z zaviralcem aromataze je obvezna ovarijska ablacija ali supresija z agonistom gonadoliberina. Hematološke bolezni: Pri nevtropeniji stopnje 3 ali 4 je priporočljiva prekinitev odmerjanja, zmanjšanje odmerka ali odložitev začetka ciklov zdravljenja, bolnike pa je treba ustrezno spremljati. ILD/pnevmonitis: Pri bolnikih se lahko pojavita huda, življenjsko ogrožajoča ali smrtna ILD in/ali pnevmonitis, kadar zdravilo jemljejo v kombinaciji z endokrinim zdravljenjem. Bolnike je treba spremljati glede pljučnih simptomov, ki kažejo na ILD/pnevmonitis (npr. hipoksija, kašelj, dispneja), in pri pojavu novih ali poslabšanju respiratornih simptomov oz. sumu na ILD/pnevmonitis zdravljenje prekiniti. Okužbe: Zdravilo lahko poveča nagnjenost k okužbam, zato je bolnike treba spremljati glede znakov in simptomov okužbe ter jih ustrezno zdraviti. Okvara jeter ali ledvic: Pri bolnikih z zmerno ali hudo okvaro jeter ali ledvic je treba zdravilo uporabljati previdno in skrbno spremljati znake toksičnosti. (1) Laktoza: Vsebuje laktozo. Bolniki z redko dedno intoleranco za galaktozo, odsotnostjo encima laktaze ali malabsorpcijo glukoze-galaktoze ne smejo jemati tega zdravila. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij: Učinki drugih zdravil na farmakokinetiko palbocikliba: Zaviralci CYP3A: Sočasni uporabi močnih zaviralcev CYP3A, med drugim klaritromicina, indinavirja, itrakonazola, ketokonazola, lopinavirja/ritonavirja, nefazodona, nelfinavirja, posakonazola, sakvinavirja, telaprevirja, telitromicina, vorikonazola in grenivke ali grenivkinega soka, se je treba izogibati. Induktorji CYP3A: Sočasni uporabi močnih induktorjev CYP3A, med drugim karbamazepina, enzalutamida, fenitoina, rifampicina in šentjanževke, se je treba izogibati. Učinek zdravil za zmanjševanje kisline: (1) Če palbociklib zaužijemo s hrano, klinično pomembnega učinka na izpostavljenost palbociklibu ni pričakovati. (2) Klinično pomembnega učinka na izpostavljenost palbociklibu ni pričakovati. Učinki palbocikliba na farmakokinetiko drugih zdravil: Pri sočasni uporabi bo morda treba zmanjšati odmerek občutljivih substratov CYP3A z ozkim terapevtskim indeksom (npr. alfentanil, ciklosporin, dihidroergotamin, ergotamin, everolimus, fentanil, pimozid, kinidin, sirolimus in takrolimus), saj IBRANCE lahko poveča izpostavljenost tem zdravilom. Študije in vitro s prenašalci: Palbociklib lahko zavira prenos, posredovan s P-gp v prebavilih in beljakovino odpornosti pri raku dojk (BCRP). Uporaba palbocikliba z zdravili, ki so substrati P-gp (npr. digoksin, dabigatran, kolhicin) ali BCRP (npr. pravastatin, rosuvastatin, sulfasalazin) lahko poveča njihov terapevtski učinek in neželene učinke. Palbociklib lahko zavira privzemni prenašalec organskih kationov OCT1. Plodnost, nosečnost in dojenje: Med zdravljenjem in vsaj 3 tedne (ženske) oziroma 14 tednov (moški) po koncu zdravljenja je treba uporabljati ustrezne kontracepcijske metode. Zdravila ne uporabljajte pri nosečnicah in ženskah v rodni dobi, ki ne uporabljajo kontracepcije. Bolnice, ki prejemajo palbociklib, ne smejo dojiti. Zdravljenje s palbociklibom lahko ogrozi plodnost pri moških. Pred začetkom zdravljenja naj moški zato razmislijo o hrambi sperme. Vpliv na sposobnost vožnje in upravljanja s stroji: Ima blag vpliv na sposobnost vožnje in upravljanja strojev. Potrebna je previdnost. Neželeni učinki: Zelo pogosti: okužbe, nevtropenija, levkopenija, anemija, trombocitopenija, pomanjkanje teka, stomatitis, navzea, diareja, bruhanje, izpuščaj, alopecija, suha koža, utrujenost, astenija, pireksija, povečane vrednosti ALT/AST. Način in režim izdaje: Rp/Spec - Predpisovanje in izdaja zdravila je le na recept zdravnika specialista ustreznega področja medicine ali od njega pooblaščenega zdravnika. Imetnik dovoljenja za promet: Pfizer Europe MA EEIG, Boulevard de la Plaine 17, 1050 Bruxelles, Belgija. Datum zadnje revizije besedila: 16.07.2021 Pred predpisovanjem se seznanite s celotnim povzetkom glavnih značilnosti zdravila. PP-IBR-EEP-0236 Datum priprave: avgust 2021. Samo za strokovno javnost. HR+/HER2- = pozitiven na hormonske receptorje in negativen na receptorje humanega epidermalnega rastnega faktorja 2. Literatura: Povzetek glavnih značilnosti zdravila Ibrance, 16.7.2021. Pfizer Luxembourg SARL, GRAND DUCHY OF LUXEMBOURG, 51, Avenue J. F. Kennedy, L-1855 Pfizer, podružnica Ljubljana, Letališka cesta 29a, Ljubljana Revolucije zahtevajo strast. M-SI-00000051(v2.0) Več kot stoletje postavljamo nove standarde v diagnostiki in zdravljenju številnih bolezni. Danes nam novi viri podatkov in napredna analitika omogočajo, da zagotovimo pravo zdravljenje za pravega bolnika ob pravem času. Zato se povezujemo s tistimi, ki stremijo k istemu cilju in razumejo, da nova znanja služijo ne samo znanosti, temveč predvsem človeštvu. Informacija pripravljena: september 2020. DODATNE INFORMACIJE SO NA VOLJO PRI: Roche farmacevtska družba d.o.o., Stegne 13g, 1000 Ljubljana. SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA ZA ZDRAVILI TAFINLAR IN MEKINIST KO PRI VAŠIH BOLNIKIH Z MELANOMOM STADIJA III ALI IV UGOTOVITE PRISOTNOST MUTACIJE BRAF ODGOVORITE S PREIZKUŠENIM OROŽJEM Dosežite podaljšano preživetje pri bolnikih z BRAF+ melanomom stadija IV ali možnost ozdravitve pri bolnikih s stadijem III s kombinacijo zdravil 3, 4 # TAFINLAR + MEKINIST. * † * V študiji COMBI-AD je bila po medianem času spremljanja 60 mesecev (dabrafenib in trametinib), oz. 58 mesecev (placebo) ocenjena stopnja ozdravitve 52 % (95-% IZ, 48 %-58 %; dabrafenib in trametinib), in 36 % (95-% IZ, 32 %-41 %; placebo). V združeni populaciji bolnikov iz študij COMBI-d in COMBI-v je bila stopnja celokupnega preživetja bolnikov v skupini zdravljeni s kombinacijo zdravil dabrafenib in trametinib po 5 letih 34 % (95-% IZ, 30 %-38 %) v primerjavi s 27% (dabrafenib+placebo) in 23 % (vemurafenib). † Zdravili TAFINLAR in MEKINIST sta v kombinaciji indicirani za zdravljenje odraslih bolnikov z inoperabilnim ali metastatskim melanomom z mutacijo BRAF V600 in adjuvantno zdravljenje odraslih bolnikov po totalni resekciji melanoma stadija III z mutacijo BRAF V600.1,2 Imena zdravil: Tafinlar 50 mg trde kapsule, Tafinlar 75 mg trde kapsule, Mekinist 0,5 mg filmsko obložene tablete, Mekinist 2 mg filmsko obložene tablete. Sestava: Ena trda kapsula zdravila Tafinlar vsebuje dabrafenibijev mesilat, ki ustreza 50 mg dabrafeniba ali 75 mg dabrafeniba. Ena filmsko obložena tableta zdravila Mekinist vsebuje 0,5 mg trametiniba ali 2 mg trametiniba v obliki trametinibijevega dimetilsulfoksida. Indikacija: Melanom: Dabrafenib in trametinib sta v kombinaciji indicirana za zdravljenje odraslih bolnikov z inoperabilnim ali metastatskim melanomom z mutacijo BRAF V600. Dabrafenib in trametinib sta oba tudi v monoterapiji indicirana za zdravljenje odraslih bolnikov z inoperabilnim ali metastatskim melanomom z mutacijo BRAF V600. Trametinib v monoterapiji ni izkazal klinične aktivnosti pri bolnikih, ki jim je bolezen napredovala med predhodnim zdravljenjem z zaviralcem BRAF. Adjuvantno zdravljenje melanoma: Dabrafenib in trametinib sta v kombinaciji indicirana za adjuvantno zdravljenje odraslih bolnikov po totalni resekciji melanoma stadija III z mutacijo BRAF V600. Nedrobnocelični pljučni rak (NDCPR): Dabrafenib in trametinib sta v kombinaciji indicirana za zdravljenje odraslih bolnikov z napredovalim nedrobnoceličnim pljučnim rakom z mutacijo BRAF V600. Odmerjanje: Zdravljenje mora uvesti in nadzorovati zdravnik, ki ima izkušnje z uporabo zdravil proti raku. Pred uporabo dabrafeniba in/ali trametiniba mora biti z validirano preiskavo potrjeno, da ima bolnik mutacijo BRAF V600. Kombinirano zdravljenje: 150 mg dabrafeniba 2x/dan in 2 mg trametiniba 1x/dan. Dabrafenib v monoterapiji (melanom): 150 mg dabrafeniba 2x/dan. Trametinib v monoterapiji (melanom): 2 mg trametiniba 1x/dan. Če bolnik pozabi vzeti odmerek trametiniba, naj ga vzame samo, če je do naslednjega rednega odmerka več kot 12 ur, pozabljenega odmerka dabrafeniba ne sme vzeti, če je do naslednjega odmerka po razporedu manj kot 6 ur. Zdravljenje je priporočljivo nadaljevati, dokler bolniku koristi oz. do pojava nesprejemljivih toksičnih učinkov. Pri adjuvantnem zdravljenju melanoma je treba bolnike zdraviti 12 mesecev, razen če pride do ponovitve bolezni ali nesprejemljivih toksičnih učinkov. Obvladovanje neželenih učinkov lahko zahteva znižanje odmerka, prekinitev zdravljenja ali prenehanje zdravljenja. Prilagoditve odmerka ali prekinitve zdravljenja niso priporočljive v primeru neželenih učinkov ploščatoceličnega karcinoma kože ali novega primarnega melanoma. Če pri uporabi kombinacije dabrafeniba in trametiniba pride do toksičnih učinkov zdravljenja, je treba sočasno znižati odmerek obeh zdravil oz. sočasno začasno prekiniti ali dokončno ukiniti obe zdravljenji. Izjeme, pri katerih je treba odmerek prilagajati samo pri enem od obeh zdravil, so pojav zvišane telesne temperature (dabrafenib), uveitisa (dabrafenib), nekožnih malignomov z mutacijo RAS (dabrafenib), zmanjšanja iztisnega deleža levega prekata (LVEF) (trametinib), zapore mrežnične vene (RVO) ali odstopa mrežničnega pigmentnega epitelija (RPED) (trametinib) in intersticijske bolezni pljuč (IBP)/pnevmonitisa (trametinib). Za natančnejša navodila glede prilagajanja odmerkov glejte povzetka glavnih značilnosti zdravil Tafinlar in Mekinist. Bolnikom z blago ali zmerno okvaro ledvic ali z blago okvaro jeter odmerkov dabrafeniba in trametiniba ni treba prilagoditi. Pri bolnikih s hudo okvaro ledvic ali z zmerno ali hudo okvaro jeter je treba dabrafenib in trametinib, bodisi v monoterapiji ali v kombinaciji, uporabljati previdno. Bolnikom, starim > 65 let, začetnega odmerka dabrafeniba in trametiniba ni treba prilagoditi, je pa pri teh bolnikih lahko potrebno pogostejše prilagajanje odmerka trametiniba. Pri bolnikih azijske rase ni potrebno prilagajati odmerkov dabrafeniba. Varnost in učinkovitost trametiniba nista ugotovljeni pri bolnikih, ki niso belci. Varnost in učinkovitost dabrafeniba in trametiniba pri otrocih in mladostnikih (< 18 let) nista bili dokazani. Način uporabe: Zdravilo Tafinlar: Kapsule je treba zaužiti cele z vodo najmanj 1 uro pred jedjo oz. najmanj 2 uri po jedi. Ne sme se jih zgristi ali odpreti. Če bolnik po zaužitju dabrafeniba ali trametiniba bruha, odmerka ne sme vzeti ponovno, temveč mora vzeti naslednji odmerek ob običajnem času. Zdravilo Mekinist: Tablete je treba zaužiti s polnim kozarcem vode vsaj 1 uro pred jedjo ali vsaj 2 uri po jedi. Ne sme se jih gristi ali drobiti. Kontraindikacije: Preobčutljivost na učinkovini ali katero koli pomožno snov. Posebna opozorila in previdnostni ukrepi: Dabrafeniba se ne sme uporabljati pri bolnikih z melanomom in pri bolnikih z NDCPR z divjim tipom BRAF. O uporabi kombinacije dabrafeniba in trametiniba pri bolnikih z melanomom, pri katerih je bolezen napredovala med predhodnim zdravljenjem z zaviralcem BRAF, je na voljo malo podatkov, ki pa kažejo, da je učinkovitost kombinacije pri teh bolnikih manjša. Ploščatocelični karcinom kože (dabrafenib ali kombinirano zdravljenje): Opisani so primeri ploščatoceličnega karcinoma kože. Priporočljivo je opraviti pregled kože pred uvedbo dabrafeniba, vsak mesec med zdravljenjem in še do 6 mesecev po zdravljenju ploščatoceličnega karcinoma kože. Bolnika se mora spremljati še 6 mesecev po prenehanju zdravljenja z dabrafenibom ali do uvedbe drugega antineoplastičnega zdravljenja. Primere ploščatoceličnega karcinoma kože je treba zdraviti z dermatološko ekscizijo, z dabrafenibom oz. kombinacijo pa nadaljevati brez prilagoditve odmerka. Bolnikom je treba naročiti, naj nemudoma obvestijo zdravnika, če se jim pojavi kakšna nova sprememba. Nov primarni melanom (dabrafenib ali kombinirano zdravljenje): Bolnika je mogoče zdraviti z ekscizijo, spremembe v zdravljenju niso potrebne. Nadzor kot pri ploščatoceličnem karcinomu kože. Nekožni malignomi (dabrafenib ali kombinirano zdravljenje): Dabrafenib lahko poveča tveganje za razvoj nekožnih malignomov, če so prisotne mutacije RAS. Bolnikom je treba pred uvedbo zdravljenja pregledati glavo in vrat (najmanj ogled ustne sluznice in palpacijo bezgavk), opraviti morajo CT prsnega koša/trebuha. Med zdravljenjem jih je treba nadzirati, kot je klinično primerno; to lahko vključuje pregled glave in vratu na 3 mesece in CT prsnega koša/trebuha na 6 mesecev. Pred zdravljenjem in na koncu zdravljenja (ter kadar koli je klinično indicirano) sta priporočljiva analni in ginekološki pregled. Opraviti je treba pregled celotne krvne slike in biokemične preiskave krvi, kot je klinično indicirano. Krvavitev (trametinib ali kombinirano zdravljenje): Prihajalo je do krvavitev, med katerimi so bile tudi večje krvavitve in krvavitve, zaradi katerih so bolniki umrli. Tveganje se lahko poveča v primeru sočasne uporabe antiagregacijskih ali antikoagulantnih zdravil. Če pride do krvavitve, je treba bolnika zdraviti v skladu s kliničnimi indikacijami. LVEF (trametinib ali kombinirano zdravljenje): Poročali so, da trametinib zmanjša LVEF. Vsem bolnikom je treba LVEF oceniti pred uvedbo trametiniba, 1 mesec po uvedbi in nato na približno 3 mesece med zdravljenjem. Pri jemanju trametiniba v kombinaciji z dabrafenibom, so poročali o akutni hudi disfunkciji levega prekata zaradi miokarditisa. Zdravniki naj bodo pozorni na možnost miokarditisa pri bolnikih, pri katerih se znaki ali simptomi težav s srcem pojavijo na novo oziroma se poslabšajo. Zvišana telesna temperatura (dabrafenib, trametinib ali kombinirano zdravljenje): Poročali so o zvišanju telesne temperature. Pri kombiniranem zdravljenju sta pogostnost in izraženost večji. Zdravljenje z dabrafenibom je treba prekiniti, če je bolnikova telesna temperatura ≥ 38,5 °C in bolnike oceniti glede znakov in simptomov okužbe. Ko mine, je mogoče zdravljenje z dabrafenibom znova začeti ob ustrezni profilaksi z uporabo nesteroidnih protivnetnih zdravil ali paracetamola. Če antipiretiki ne zadoščajo, je treba razmisliti o uporabi peroralnih kortikosteroidov. Če je zvišana telesna temperatura povezana z drugimi hudimi znaki ali simptomi, je treba zdravljenje z dabrafenibom znova začeti z nižjim odmerkom, ko zvišana telesna temperatura mine in kot je klinično primerno. Hipertenzija (trametinib ali kombinirano zdravljenje): Opisana so zvišanja krvnega tlaka tako pri bolnikih, ki so že prej imeli hipertenzijo, kot pri tistih, ki je prej niso imeli. Krvni tlak je treba izmeriti izhodiščno in med zdravljenjem; hipertenzijo je treba obvladovati s standardnim zdravljenjem. IBP (trametinib ali kombinirano zdravljenje): V primeru suma na IBP ali pnevmonitis je treba zdravljenje s trametinibom prekiniti; tudi pri bolnikih z novonastalimi ali napredujočimi pljučnimi simptomi ali izvidi, vključno s kašljem, dispnejo, hipoksijo, plevralnim izlivom ali infiltrati, dokler niso opravljene klinične preiskave. Pri bolnikih, ki imajo diagnosticirano z zdravljenjem povezano IBP ali pnevmonitis, je treba zdravljenje s trametinibom trajno končati. Okvara vida (trametinib ali kombinirano zdravljenje): Lahko pride do težav, povezanih z motnjami vida, vključno z RPED in RVO. Uporaba trametiniba ni priporočljiva pri bolnikih, ki so v preteklosti že imeli RVO. Če se pojavijo novonastale motnje vida, npr. poslabšanje centralnega vida, zamegljen vid ali izguba vida, je priporočljiva takojšnja oftalmološka ocena. Pri bolnikih z diagnozo RVO, je treba trametinib trajno ukiniti. Okvara vida (dabrafenib ali kombinirano zdravljenje): Opisovali so oftalmološke reakcije, vključno z uveitisom, iridociklitisom in/ali iritisom. Bolnike je treba redno kontrolirati glede znakov in simptomov s strani vida (npr. sprememb vida, fotofobije in bolečin v očesu). Prilagajanje odmerka ni potrebno, dokler je očesno vnetje mogoče obvladati z učinkovitimi lokalnimi zdravili. Če se uveitis ne odziva na lokalna očesna zdravila, je treba zdravljenje z dabrafenibom prekiniti, dokler očesno vnetje ni odpravljeno, nato pa ga ponovno uvesti v odmerku, ki je za eno odmerno raven nižji. Odpoved ledvic (dabrafenib ali kombinirano zdravljenje): Med zdravljenjem je treba rutinsko določati vrednost kreatinina v serumu. Če se vrednost zviša, je morda treba začasno prekiniti uporabo dabrafeniba, če je to klinično primerno. Uporabe dabrafeniba niso preučili pri bolnikih z insuficienco ledvic (kreatinin > 1,5 x ZMN), zato ga je treba v takšnih okoliščinah uporabljati previdno. Pankreatitis (dabrafenib ali kombinirano zdravljenje): Poročali so o primerih pankreatitisa. Nepojasnjene bolečine v trebuhu je treba nemudoma raziskati, vključno z meritvijo amilaze in lipaze v serumu. Ob ponovnem začetku uporabe dabrafeniba po pankreatitisu je treba bolnike skrbno kontrolirati. Jetrni dogodki (trametinib ali kombinirano zdravljenje): Prvih 6 mesecev po začetku zdravljenja s trametinibom je delovanje jeter priporočljivo kontrolirati na 4 tedne in nato kot je klinično indicirano. Globoka venska tromboza/pljučna embolija (trametinib ali kombinirano zdravljenje): Če se pri bolniku pojavijo znaki pljučne embolije ali globoke venske tromboze, kot so zadihanost, bolečine v prsnem košu ali zatekanje rok ali nog, mora takoj poiskati zdravniško pomoč. Če gre za življenjsko nevarno pljučno embolijo, je treba bolniku dokončno ukiniti zdravljenje s trametinibom in dabrafenibom. Hude kožne neželene reakcije: Pred uvedbo zdravljenja je treba bolnike opozoriti na znake in simptome kožnih reakcij in jih skrbno spremljati. Če se pojavijo znaki in simptomi, ki lahko pomenijo, da gre za hudo kožno neželeno reakcijo, mora bolnik prekiniti zdravljenje z dabrafenibom in trametinibom. Bolezni prebavil (trametinib ali kombinirano zdravljenje): Poročali so o kolitisu in perforaciji prebavil, vključno s primeri, ki so se končali s smrtjo. Previdnost je potrebna pri uporabi trametiniba pri bolnikih z dejavniki tveganja za perforacijo prebavil, kot so divertikulitis v anamnezi, metastaze v prebavnem traktu ali sočasna uporaba zdravil z znanim tveganjem za perforacijo prebavil. Sarkoidoza: Ob diagnozi sarkoidoze je treba razmisliti o ustreznem zdravljenju. Pomembno je, da se sarkoidoza ne interpretira kot napredovanje bolezni. Plodnost, nosečnost in dojenje: Ženske v rodni dobi morajo uporabljati učinkovite kontracepcijske metode med zdravljenjem in še 2 tedna po prenehanju zdravljenja z dabrafenibom ter še 16 tednov po zadnjem odmerku trametiniba. Dabrafenib lahko zmanjša učinkovitost peroralnih oz. katerihkoli hormonskih kontraceptivov, zato je treba uporabiti drug učinkovit način kontracepcije. Trenutno ni znano, ali trametinib vpliva na hormonske kontraceptive. Za preprečitev nosečnosti se bolnicam, ki jemljejo hormonske kontraceptive svetuje, da uporabijo dodaten ali drug način kontracepcije med zdravljenjem in še 16 tednov po prekinitvi zdravljenja s trametinibom. Nosečnice in doječe matere ne smejo dobivati trametiniba. Nosečnice naj ne bi prejemale dabrafeniba, razen če možna korist za mater odtehta možno tveganje za plod. Odločiti se je treba bodisi za prenehanje dojenja bodisi za prenehanje zdravljenja z dabrafenibom, upoštevaje koristi dojenja za otroka in koristi zdravljenja za žensko. Dabrafenib in trametinib lahko prizadeneta plodnost moških in žensk. Moške bolnike, ki jemljejo dabrafenib in/ali trametinib, je treba seznaniti z možnim tveganjem za motnje spermatogeneze, ki so lahko ireverzibilne. Dabrafenib in trametinib imata blag vpliv na sposobnost vožnje in upravljanja strojev. Bolnika je treba seznaniti z možnostjo za utrujenost, omotico in težave z očmi, ki lahko vplivajo na takšne dejavnosti. Medsebojno delovanje z drugimi zdravili: Zdravilo Tafinlar: Verjetno je, da zdravila, ki močno zavirajo ali inducirajo CYP2C8 ali CYP3A4, povečajo oz. zmanjšajo koncentracijo dabrafeniba. Če je mogoče, je treba med uporabo dabrafeniba uporabiti druga zdravila. V primeru uporabe dabrafeniba z močnimi zaviralci (npr. ketokonazolom, gemfibrozilom, nefazodonom, klaritromicinom, ritonavirjem, sakvinavirjem, telitromicinom, itrakonazolom, vorikonazolom, posakonazolom, atazanavirjem) je potrebna previdnost. Izogibajte se sočasni uporabi dabrafeniba z močnimi induktorji (npr. rifampicinom, fenitoinom, karbamazepinom, fenobarbitalom ali šentjanževko (Hypericum perforatum)) CYP2C8 ali CYP3A4. Ne pričakuje se, da bi zdravila, ki spreminjajo pH zgornjega gastrointestinalnega trakta (npr. zaviralci protonske črpalke, antagonisti histaminskih receptorjev H2, antacidi), zmanjšala biološko uporabnost dabrafeniba. Dabrafenib je induktor encimov in poveča sintezo encimov, ki presnavljajo zdravila. Posledica je manjša plazemska koncentracija zdravil, ki se presnavljajo s temi encimi, kar lahko povzroči izgubo ali zmanjšanje kliničnega učinka teh zdravil. Pričakovati je mogoče medsebojna delovanja s številnimi zdravili, ki se izločajo s presnavljanjem ali aktivnim transportom, a velikost medsebojnega delovanja se razlikuje. To lahko velja za, vendar ni omejeno na naštete skupine zdravil: analgetiki (npr. fentanil, metadon), antibiotiki (npr. klaritromicin, doksiciklin), zdravila proti raku (npr. kabazitaksel), antikoagulansi (npr. acenokumarol, varfarin), antiepileptiki (npr. karbamazepin, fenitoin, primidon, valprojska kislina), antipsihotiki (npr. haloperidol), zaviralci kalcijevih kanalčkov (npr. diltiazem, felodipin, nikardipin, nifedipin, verapamil), srčni glikozidi (npr. digoksin), kortikosteroidi (npr. deksametazon, metilprednizolon), protivirusna zdravila proti HIV (npr. amprenavir, atazanavir, darunavir, delavirdin, efavirenz, fosamprenavir, indinavir, lopinavir, nelfinavir, sakvinavir, tipranavir), hormonski kontraceptivi, hipnotiki (npr. diazepam, midazolam, zolpidem), imunosupresivi (npr. ciklosporin, takrolimus, sirolimus), statini, ki se presnavljajo s CYP3A4 (npr. atorvastatin, simvastatin). Če je njihov terapevtski učinek za bolnika zelo pomemben in če odmerka ni mogoče zlahka prilagoditi glede na kontrolo učinkovitosti ali koncentracije v plazmi, se je tem zdravilom treba izogniti ali jih je treba uporabljati previdno. Tveganje za okvaro jeter po uporabi paracetamola je domnevno večje pri bolnikih, ki sočasno prejemajo induktorje encimov. Pojav indukcije je verjeten po 3 dneh ponavljajočega se odmerjanja dabrafeniba. Po prenehanju uporabe dabrafeniba indukcija mine postopoma. Bolnike je treba spremljati glede toksičnih učinkov; potrebna je lahko prilagoditev odmerjanja navedenih zdravil. Zdravilo Mekinist: Zaradi možnosti zvišanja koncentracije trametiniba, je priporočena previdnost pri sočasnem odmerjanju trametiniba in zdravil, ki so močni zaviralci P‑gp (na primer verapamila, ciklosporina, ritonavirja, kinidina, itrakonazola). Trametinib lahko povzroči prehodno zavrtje substratov BCRP (npr. pitavastatina) v črevesu; to je mogoče omejiti na najmanjšo mero tako, da se ta zdravila in trametinib uporablja z medsebojnim zamikom (zamik 2 ur). Neželeni učinki: Dabrafenib v monoterapiji: Zelo pogosti (≥ 1/10): papilom, zmanjšan apetit, glavobol, kašelj, navzea, bruhanje, driska, hiperkeratoza, alopecija, izpuščaj, sindrom palmarno-plantarne eritrodisestezije, artralgija, mialgija, bolečine v okončini, pireksija, utrujenost, mrzlica, astenija. Pogosti (≥ 1/100 do < 1/10): ploščatocelični karcinom kože, seboroična keratoza, akrohordon (kožni izrastki), bazalnocelični karcinom, hipofosfatemija, hiperglikemija, zaprtost, suha koža, srbenje, aktinična keratoza, kožne lezije, eritem, fotosenzitivnost, gripi podobna bolezen. Občasni (≥ 1/1.000 do < 1/100): nov primarni melanom, preobčutljivost, uveitis, pankreatitis, panikulitis, odpoved ledvic, akutna odpoved ledvic, nefritis. Trametinib v monoterapiji: Zelo pogosti: hipertenzija, krvavitev, kašelj, dispneja, driska, navzea, bruhanje, zaprtost, bolečine v trebuhu, suha usta, izpuščaj, akneiformni dermatitis, suha koža, srbenje, alopecija, utrujenost, periferni edemi, zvišana telesna temperatura, zvišana aspartat-aminotransferaza. Pogosti: folikulitis, paronihija, celulitis, pustulozen izpuščaj, anemija, preobčutljivost, dehidracija, zamegljen vid, periorbitalni edem, okvara vida, disfunkcija levega prekata, zmanjšanje iztisnega deleža, bradikardija, limfedem, pnevmonitis, stomatitis, eritem, sindrom palmarno-plantarne eritrodisestezije, fisure na koži, razpokana koža, edem obraza, vnetje sluznice, astenija, zvišana alanin-aminotransferaza, zvišana alkalna fosfataza v krvi, zvišana kreatin-fosfokinaza v krvi. Občasni: horioretinopatija, papiledem, odstop mrežnice, zapora mrežnične vene, srčno popuščanje, intersticijska bolezen pljuč, perforacija prebavil, kolitis, rabdomioliza. Kombinirano zdravljenje z dabrafenibom in trametinibom: Zelo pogosti:nazofaringitis, zmanjšan apetit, glavobol, omotica, hipertenzija, krvavitev, kašelj, bolečine v trebuhu, zaprtost, diareja, navzea, bruhanje, suha koža, srbenje, izpuščaj, eritem, artralgija, mialgija, bolečine v okončini, mišični krči, utrujenost, mrzlica, astenija, periferni edemi, zvišana telesna temperatura, gripi podobna bolezen, zvišana vrednost alanin aminotransferaze, zvišana vrednost aspartat aminotransferaze. Pogosti: okužba sečil, celulitis, folikulitis, paronihija, pustulozen izpuščaj, ploščatocelični karcinom kože, papilom, seboroična keratoza, nevtropenija, anemija, trombocitopenija, levkopenija, dehidracija, hiponatriemija, hipofosfatemija, hiperglikemija, zamegljen vid, okvara vida, uveitis, zmanjšanje iztisnega deleža, limfedem, hipotenzija, dispneja, suha usta, stomatitis, akneiformni dermatitis, aktinična keratoza, nočno znojenje, hiperkeratoza, alopecija, sindrom palmarno-plantarne eritrodisestezije, kožne spremembe, čezmerno znojenje, panikulitis, fisure na koži, fotosenzitivnost, vnetje sluznice, edem obraza, zvišana vrednost alkalne fosfataze v krvi, zvišana vrednost gama-glutamiltransferaze, zvišana vrednost kreatin-fosfokinaze v krvi. Občasni: nov primarni melanom, akrohordon (pecljati fibrom), preobčutljivost, sarkoidoza, horioretinopatija, odstop mrežnice, periorbitalni edem, bradikardija, pnevmonitis, pankreatitis, kolitis, odpoved ledvic, nefritis. Redki: perforacija prebavil. Neznana pogostnost: miokarditis, Stevens‑Johnsonov sindrom, reakcija na zdravilo z eozinofilijo in sistemskimi simptomi (DRESS sindrom), generaliziran eksfoliativni dermatitis. Imetnik dovoljenja za promet z zdravilom: Novartis Europharm Limited, Vista Building, Elm Park, Merrion Road, Dublin 4, Irska Dodatne informacije in literatura: Novartis Pharma Services Inc., Podružnica v Sloveniji, Verovškova ulica 57, 1000 Ljubljana. Način/režim izdajanja zdravil Tafinlar in Mekinist: Rp/Spec. Pred predpisovanjem natančno preberite zadnji odobreni povzetek glavnih značilnosti zdravila. Datum zadnje revizije skrajšanega povzetka glavnih značilnosti zdravila: januar 2021. Viri in literatura: 1. Povzetek glavnih značilnosti zdravila Tafinlar. Januar 2021. 2. Povzetek glavnih značilnosti zdravila Mekinist. Januar 2021. 3. Dummer R, Hauschild A, Santinami M, et al. Five-Year Analysis of Adjuvant Dabrafenib plus Trametinib in Stage III Melanoma. N Engl J Med 2020;383:1139-48. 4. Robert C, Grob JJ, Stroyakovskiy D, et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N Engl J Med 2019;381:626-36. # Novartis Pharma Services Inc., Podružnica v Sloveniji Verovškova ulica 57, 1000 Ljubljana, tel.: 01 300 75 50 Datum priprave materiala: februar 2021 Samo za strokovno javnost SI-2021-MEK-014 POWERING TARGETED THERAPY Decades of innovation in targeted therapies is helping people with lung cancer live longer © 2020 Amgen Inc. All rights reserved. Amgen zdravila d.o.o., Ameriška ulica 2, Ljubljana Date of preparation: August 2021. SI-510-0821-00001 vsak dan abemaciklib dvakrat na dan EDINI zaviralec CDK4 & 6, ki se jemlje NEPREKINJENO VSAK DAN, NE 2x NA DAN1, 2, 3 SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA Za to zdravilo se izvaja dodatno spremljanje varnosti. Tako bodo hitreje na voljo nove informacije o njegovi varnosti. Zdravstvene delavce naprošamo, da poročajo o katerem koli domnevnem neželenem učinku zdravila. Glejte poglavje 4.8, kako poročati o neželenih učinkih. IME ZDRAVILA: Verzenios 50 mg/100 mg/150 mg filmsko obložene tablete KAKOVOSTNA IN KOLIČINSKA SESTAVA: Ena filmsko obložena tableta vsebuje 50 mg/100 mg/150 mg abemacikliba. Ena filmsko obložena tableta vsebuje 14 mg/28 mg/42 mg laktoze (v obliki monohidrata). Terapevtske indikacije: Zdravilo Verzenios je indicirano za zdravljenje žensk z lokalno napredovalim ali metastatskim, na hormonske receptorje (HR – Hormone Receptor) pozitivnim in na receptorje humanega epidermalnega rastnega faktorja 2 (HER2 – Human Epidermal Growth Factor Receptor 2) negativnim rakom dojk v kombinaciji z zaviralcem aromataze ali s fulvestrantom kot začetnim endokrinim zdravljenjem ali pri ženskah, ki so prejele predhodno endokrino zdravljenje. Pri ženskah v pred- in perimenopavzi je treba endokrino zdravljenje kombinirati z agonistom gonadoliberina (LHRH – Luteinizing Hormone–Releasing Hormone). Odmerjanje in način uporabe: Zdravljenje z zdravilom Verzenios mora uvesti in nadzorovati zdravnik, ki ima izkušnje z uporabo zdravil za zdravljenje rakavih bolezni. Zdravilo Verzenios v kombinaciji z endokrinim zdravljenjem: Priporočeni odmerek abemacikliba je 150 mg dvakrat na dan, kadar se uporablja v kombinaciji z endokrinim zdravljenjem. Zdravilo Verzenios je treba jemati, dokler ima bolnica od zdravljenja klinično korist ali do pojava nesprejemljive toksičnosti. Če bolnica bruha ali izpusti odmerek zdravila Verzenios, ji je treba naročiti, da naj naslednji odmerek vzame ob predvidenem času; dodatnega odmerka ne sme vzeti. Obvladovanje nekaterih neželenih učinkov lahko zahteva prekinitev in/ali zmanjšanje odmerka. Zdravljenje z abemaciklibom prekinite v primeru povišanja vrednosti AST in/ali ALT >3 x ZMN SKUPAJ s celokupnim bilirubinom > 2,0 x ZMN v odsotnosti holestaze ter pri bolnicah z intersticijsko pljučno boleznijo (ILD)/pnevmonitis stopnje 3 ali 4. Sočasni uporabi močnih zaviralcev CYP3A4 se je treba izogibati. Če se uporabi močnih zaviralcev CYP3A4 ni mogoče izogniti, je treba odmerek abemacikliba znižati na 100 mg dvakrat na dan. Pri bolnicah, pri katerih je bil odmerek znižan na 100 mg abemacikliba dvakrat na dan in pri katerih se sočasnemu dajanju močnega zaviralca CYP3A4 ni mogoče izogniti, je treba odmerek abemacikliba dodatno znižati na 50 mg dvakrat na dan. Pri bolnicah, pri katerih je bil odmerek znižan na 50 mg abemacikliba dvakrat na dan in pri katerih se sočasnemu dajanju močnega zaviralca CYP3A4 ni mogoče izogniti, je mogoče z odmerkom abemacikliba nadaljevati ob natančnem spremljanju znakov toksičnosti. Alternativno je mogoče odmerek abemacikliba znižati na 50 mg enkrat na dan ali prekiniti dajanje abemacikliba. Če je uporaba zaviralca CYP3A4 prekinjena, je treba odmerek abemacikliba povečati na odmerek, kakršen je bil pred uvedbo zaviralca CYP3A4 (po 3–5 razpolovnih časih zaviralca CYP3A4). Prilagajanje odmerka glede na starost in pri bolnicah z blago ali zmerno ledvično okvaro ter z blago (Child Pugh A) ali zmerno (Child Pugh B) jetrno okvaro ni potrebno. Pri dajanju abemacikliba bolnicam s hudo ledvično okvaro sta potrebna previdnost in skrbno spremljanje glede znakov toksičnosti. Način uporabe: Zdravilo Verzenios je namenjeno za peroralno uporabo. Odmerek se lahko vzame s hrano ali brez nje. Zdravilo se ne sme jemati z grenivko ali grenivkinim sokom. Bolnice naj odmerke vzamejo vsak dan ob približno istem času. Tableto je treba zaužiti celo (bolnice je pred zaužitjem ne smejo gristi, drobiti ali deliti). Kontraindikacije: Preobčutljivost na učinkovino ali katero koli pomožno snov. Posebna opozorila in previdnostni ukrepi: Pri bolnicah, ki so prejemale abemaciklib, so poročali o nevtropeniji, o večji pogostnosti okužb kot pri bolnicah, zdravljenih s placebom in endokrinim zdravljenjem, o povečanih vrednostih ALT in AST. Pri bolnicah, pri katerih se pojavi nevtropenija stopnje 3 ali 4, je priporočljivo prilagoditi odmerek. Bolnice je treba spremljati za znake in simptome globoke venske tromboze in pljučne embolije ter jih zdraviti, kot je medicinsko utemeljeno. Glede na povečanje vrednosti ALT ali AST je mogoče potrebna prilagoditev odmerka. Driska je najpogostejši neželeni učinek. Bolnice je treba ob prvem znaku tekočega blata začeti zdraviti z antidiaroiki, kot je loperamid, povečati vnos peroralnih tekočin in obvestiti zdravnika. Sočasni uporabi induktorjev CYP3A4 se je treba izogibati zaradi tveganja za zmanjšano učinkovitost abemacikliba. Bolnice z redkimi dednimi motnjami, kot so intoleranca za galaktozo, popolno pomanjkanje laktaze ali malapsorpcija glukoze/galaktoze, tega zdravila ne smejo jemati. Bolnice spremljajte glede pljučnih simptomov, ki kažejo na ILD/pnevmonitis, in jih ustrezno zdravite. Glede na stopnjo ILD/pnevmonitisa je morda potrebno prilagajanje odmerka abemacikliba. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij: Abemaciklib se primarno presnavlja s CYP3A4. Sočasna uporaba abemacikliba in zaviralcev CYP3A4 lahko poveča plazemsko koncentracijo abemacikliba. Uporabi močnih zaviralcev CYP3A4 sočasno z abemaciklibom se je treba izogibati. Če je močne zaviralce CYP3A4 treba dajati sočasno, je treba odmerek abemacikliba zmanjšati, nato pa bolnico skrbno spremljati glede toksičnosti. Pri bolnicah, zdravljenih z zmernimi ali šibkimi zaviralci CYP3A4, ni potrebno prilagajanje odmerka, vendar jih je treba skrbno spremljati za znake toksičnosti. Sočasni uporabi močnih induktorjev CYP3A4 (vključno, vendar ne omejeno na: karbamazepin, fenitoin, rifampicin in šentjanževko) se je treba izogibati zaradi tveganja za zmanjšano učinkovitost abemacikliba. Abemaciklib in njegovi glavni aktivni presnovki zavirajo prenašalce v ledvicah, in sicer kationski organski prenašalec 2 (OCT2) ter prenašalca MATE1. In vivo lahko pride do medsebojnega delovanja abemacikliba in klinično pomembnih substratov teh prenašalcev, kot je dofelitid ali kreatinin. Trenutno ni znano, ali lahko abemaciklib zmanjša učinkovitost sistemskih hormonskih kontraceptivov, zato se ženskam, ki uporabljajo sistemske hormonske kontraceptive, svetuje, da hkrati uporabljajo tudi mehansko metodo. Neželeni učinki: Najpogostejši neželeni učinki so driska, okužbe, nevtropenija, anemija, utrujenost, navzea, bruhanje in zmanjšanje apetita. Zelo pogosti: okužbe, nevtropenija, levkopenija, anemija, trombocitopenija, driska, bruhanje, navzea, zmanjšanje apetita, disgevzija, omotica, alopecija, pruritus, izpuščaj, utrujenost, pireksija, povečana vrednost alanin-aminotransferaze, povečana vrednost aspartat-aminotransferaze Pogosti: limfopenija, povečano solzenje, venska trombembolija, intersticijska pljučna bolezen (ILD)/pnevmonitis, suha koža, mišična šibkost Občasni: febrilna nevtropenija Rok uporabnosti: 3 leta Posebna navodila za shranjevanje: Za shranjevanje zdravila niso potrebna posebna navodila. Imetnik dovoljenja za promet z zdravilom: Eli Lilly Nederland B.V., Papendorpseweg 83, 3528BJ, Utrecht, Nizozemska. Datum prve odobritve dovoljenja za promet: 27. september 2018 Datum zadnje revizije besedila: 19.7.2021 Režim izdaje: Rp/Spec - Predpisovanje in izdaja zdravila je le na recept zdravnika specialista ustreznega področja medicine ali od njega pooblaščenega zdravnika. Reference: 1. Povzetek glavnih značilnosti zdravila Verzenios. Datum zadnje revizije besedila: 19.7.2021. 2. Povzetek glavnih značilnosti zdravila Ibrance. Dostop preverjen 10.4.2020. 3. Povzetek glavnih značilnosti zdravila Kisqali. Dostop preverjen 10.4.2020. Pomembno: Predpisovanje in izdaja zdravila je le na recept zdravnika specialista ustreznega področja medicine ali od njega pooblaščenega zdravnika. Pred predpisovanjem zdravila Verzenios si preberite zadnji veljavni Povzetek glavnih značilnosti zdravil. Podrobne informacije o zdravilu so objavljene na spletni strani Evropske agencije za zdravila http://www.ema.europa.eu Eli Lilly farmacevtska družba, d.o.o., Dunajska cesta 167, 1000 Ljubljana, telefon 01 / 580 00 10, faks 01 / 569 17 05 PP-AL-SI-0110, 23.8.2021, Samo za strokovno javnost. Bolniki morajo imeti dostop do zdravil, ki jih potrebujejo. podobna biološka zdravila brez meja SKRBIMO ZA VAS Peter, 42 Slovenija električar rak BIO0492020 Emmanuel, 54 Nigerija policist Crohnova bolezen Mylan Healthcare, farmacevtsko podjetje, d.o.o., Dolenjska cesta 242c, 1000 Ljubljana, www.v iatris.com/sl-si Družba Viatris po celem svetu zdravnikom in bolnikom zagotavlja dostop do visokokakovostnih bioloških zdravil. Slednjega zagotavljajo zavezanost h kontinuiranemu razvoju, kakovostnim raziskavam, nadzoru kakovosti in logistični odličnosti. To je naš svet. Naš svet je svet, ki mu je mar za vas. Strokovna knjižnica za onkologijo 8 čitalniških mest 5.300 knjig 6.000 e-revij vsak delovni dan od 8. do 15. ure www.onko-i.si/strokovna_knjiznica #2 INTERNATIONAL SUMMER SCHOOL SO PODPRLE NASLEDNJE DRUŽBE: NOVARTIS – ZLATI SPONZOR MSD – ZLATI SPONZOR SERVIER ELI LILLY PFIZER AMGEN BRISTOL-MYERS SQUIBB ROCHE EWOPHARMA LEK ABBOTT JANSSEN SANOFI GENZYME PHARMASWISS TAKEDA MYLAN MERCK