51
|
Larkin J, Del Vecchio M, Mandalá M, Gogas H, Arance Fernandez AM, Dalle S, Cowey CL, Schenker M, Grob JJ, Chiarion-Sileni V, Marquez-Rodas I, Butler MO, Di Giacomo AM, Middleton MR, Lutzky J, de la Cruz-Merino L, Arenberger P, Atkinson V, Hill AG, Fecher LA, Millward M, Nathan PD, Khushalani NI, Queirolo P, Ritchings C, Lobo M, Askelson M, Tang H, Dolfi S, Ascierto PA, Weber J. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III/IV Melanoma: 5-Year Efficacy and Biomarker Results from CheckMate 238. Clin Cancer Res 2023; 29:3352-3361. [PMID: 37058595 PMCID: PMC10472092 DOI: 10.1158/1078-0432.ccr-22-3145] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/31/2023] [Accepted: 04/12/2023] [Indexed: 04/16/2023]
Abstract
PURPOSE In the phase III CheckMate 238 study, adjuvant nivolumab significantly improved recurrence-free survival (RFS) and distant metastasis-free survival versus ipilimumab in patients with resected stage IIIB-C or stage IV melanoma, with benefit sustained at 4 years. We report updated 5-year efficacy and biomarker findings. PATIENTS AND METHODS Patients with resected stage IIIB-C/IV melanoma were stratified by stage and baseline programmed death cell ligand 1 (PD-L1) expression and received nivolumab 3 mg/kg every 2 weeks or ipilimumab 10 mg/kg every 3 weeks for four doses and then every 12 weeks, both intravenously for 1 year until disease recurrence, unacceptable toxicity, or withdrawal of consent. The primary endpoint was RFS. RESULTS At a minimum follow-up of 62 months, RFS with nivolumab remained superior to ipilimumab (HR = 0.72; 95% confidence interval, 0.60-0.86; 5-year rates of 50% vs. 39%). Five-year distant metastasis-free survival (DMFS) rates were 58% with nivolumab versus 51% with ipilimumab. Five-year overall survival (OS) rates were 76% with nivolumab and 72% with ipilimumab (75% data maturity: 228 of 302 planned events). Higher levels of tumor mutational burden (TMB), tumor PD-L1, intratumoral CD8+ T cells and IFNγ-associated gene expression signature, and lower levels of peripheral serum C-reactive protein were associated with improved RFS and OS with both nivolumab and ipilimumab, albeit with limited clinically meaningful predictive value. CONCLUSIONS Nivolumab is a proven adjuvant treatment for resected melanoma at high risk of recurrence, with sustained, long-term improvement in RFS and DMFS compared with ipilimumab and high OS rates. Identification of additional biomarkers is needed to better predict treatment outcome. See related commentary by Augustin and Luke, p. 3253.
Collapse
Affiliation(s)
- James Larkin
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | | | - Helen Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | | | | | - Ivan Marquez-Rodas
- General University Hospital Gregorio Marañón and CIBERONC, Madrid, Spain
| | | | | | | | - Jose Lutzky
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Luis de la Cruz-Merino
- Hospital Universitario Virgen Macarena, Clinical Oncology Department, University of Seville, Seville, Spain
| | - Petr Arenberger
- Charles University Third Faculty of Medicine and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Victoria Atkinson
- Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
| | | | | | - Michael Millward
- University of Western Australia and Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | | | | | | | | | | | | | - Hao Tang
- Bristol Myers Squibb, Princeton, New Jersey
| | | | - Paolo A. Ascierto
- Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York
| |
Collapse
|
52
|
Sangro B, Yau T, El‐Khoueiry AB, Kudo M, Shen Y, Tschaika M, Roy A, Feng Y, Gao L, Aras U. Exposure-response analysis for nivolumab plus ipilimumab combination therapy in patients with advanced hepatocellular carcinoma (CheckMate 040). Clin Transl Sci 2023; 16:1445-1457. [PMID: 37165980 PMCID: PMC10432868 DOI: 10.1111/cts.13544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023] Open
Abstract
This analysis was conducted to inform dose selection of a combination of nivolumab plus ipilimumab for the treatment of sorafenib-experienced patients with hepatocellular carcinoma (HCC). CheckMate 040 is an open-label, multicohort, phase I/II trial in adults with advanced HCC that evaluated nivolumab monotherapy (0.1-10 mg/kg once every 2 weeks [q2w]) and the following three combinations of nivolumab plus ipilimumab: (1) nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks (q3w) for four doses, followed by nivolumab monotherapy 240 mg q2w (arm A); (2) nivolumab 3 mg/kg plus ipilimumab 1 mg/kg q3w for four doses, followed by nivolumab monotherapy 240 mg q2w (arm B); and (3) nivolumab 3 mg/kg q2w plus ipilimumab 1 mg/kg every 6 weeks continuously (arm C). Exposure-response relationships (efficacy and safety) were characterized using nivolumab and ipilimumab concentrations after the first dose (Cavg1) as the exposure measure. Objective tumor response (OTR) and overall survival (OS) improvements were associated with increased ipilimumab exposure (OTR: odds ratio 1.45, 95% confidence interval [CI], 1.13-1.86; OS: hazard ratio 0.86, 95% CI 0.75-0.98), but not nivolumab exposure (OTR: odds ratio 0.99, 95% CI 0.97-1.02; OS: hazard ratio 1.08, 95% CI 0.89-1.32). Hepatic treatment-related and immune-mediated adverse events were more common in arm A than in arms B or C. Nivolumab 1 mg/kg plus ipilimumab 3 mg/kg q3w for four doses, followed by nivolumab monotherapy 240 mg q2w had the most favorable benefit:risk profile in patients with advanced HCC.
Collapse
Affiliation(s)
- Bruno Sangro
- Liver UnitClinica Universidad de Navarra‐IDISNA and CIBEREHDPamplonaSpain
| | - Thomas Yau
- University of Hong Kong, Hong Kong Special Administrative RegionPokfulamChina
| | | | - Masatoshi Kudo
- Department of Gastroenterology and HepatologyKindai University Faculty of MedicineOsakaJapan
| | - Yun Shen
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | | | - Amit Roy
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Yan Feng
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Ling Gao
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Urvi Aras
- Bristol Myers SquibbPrincetonNew JerseyUSA
| |
Collapse
|
53
|
Long GV, Swetter SM, Menzies AM, Gershenwald JE, Scolyer RA. Cutaneous melanoma. Lancet 2023:S0140-6736(23)00821-8. [PMID: 37499671 DOI: 10.1016/s0140-6736(23)00821-8] [Citation(s) in RCA: 274] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/17/2023] [Accepted: 04/19/2023] [Indexed: 07/29/2023]
Abstract
Cutaneous melanoma is a malignancy arising from melanocytes of the skin. Incidence rates are rising, particularly in White populations. Cutaneous melanoma is typically driven by exposure to ultraviolet radiation from natural sunlight and indoor tanning, although there are several subtypes that are not related to ultraviolet radiation exposure. Primary melanomas are often darkly pigmented, but can be amelanotic, with diagnosis based on a combination of clinical and histopathological findings. Primary melanoma is treated with wide excision, with margins determined by tumour thickness. Further treatment depends on the disease stage (following histopathological examination and, where appropriate, sentinel lymph node biopsy) and can include surgery, checkpoint immunotherapy, targeted therapy, or radiotherapy. Systemic drug therapies are recommended as an adjunct to surgery in patients with resectable locoregional metastases and are the mainstay of treatment in advanced melanoma. Management of advanced melanoma is complex, particularly in those with cerebral metastasis. Multidisciplinary care is essential. Systemic drug therapies, particularly immune checkpoint inhibitors, have substantially increased melanoma survival following a series of landmark approvals from 2011 onward.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Mater Hospital, Sydney, NSW, Australia.
| | - Susan M Swetter
- Department of Dermatology and Pigmented Lesion and Melanoma Program, Stanford University Medical Center and Cancer Institute, Stanford, CA, USA; Department of Dermatology, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Mater Hospital, Sydney, NSW, Australia
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology and Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard A Scolyer
- Melanoma Institute Australia, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; NSW Health Pathology, Sydney, NSW, Australia
| |
Collapse
|
54
|
Reyes A, Pharaon R, Mohanty A, Massarelli E. Arising Novel Agents in Lung Cancer: Are Bispecifics and ADCs the New Paradigm? Cancers (Basel) 2023; 15:3162. [PMID: 37370772 PMCID: PMC10296730 DOI: 10.3390/cancers15123162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Lung cancer is one of the most common cancers with the highest mortality. Non-small cell lung cancer (NSCLC) contributes to around 85% of lung cancer diagnoses (vs. 15% for small cell lung cancer). The treatment of NSCLC has vastly changed in the last two decades since the development of immunotherapy and targeted therapy against driver mutations. As is the nature of malignancy, cancer cells have acquired resistance to these treatments prompting an investigation into novel treatments and new targets. Bispecific antibodies, capable of targeting multiple substrates at once, and antibody-drug conjugates that can preferentially deliver chemotherapy to tumor cells are examples of this innovation. From our initial evaluation, both treatment modalities appear promising.
Collapse
Affiliation(s)
| | | | | | - Erminia Massarelli
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 910102, USA; (A.R.); (R.P.); (A.M.)
| |
Collapse
|
55
|
Faleck DM, Dougan M, Tello M, Grossman JE, Moss AC, Postow MA. Accelerating the Evolution of Immune-Related Enterocolitis Management. J Clin Oncol 2023; 41:3110-3115. [PMID: 37040601 PMCID: PMC10256374 DOI: 10.1200/jco.22.02914] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/14/2023] [Accepted: 03/09/2023] [Indexed: 04/13/2023] Open
Affiliation(s)
- David M. Faleck
- Gastroenterology, Hepatology & Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Michael Dougan
- Division of Gastroenterology and Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA
| | | | | | - Alan C. Moss
- Division of Gastroenterology, Department of Medicine, Boston Medical Center, Boston, MA
| | - Michael A. Postow
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
56
|
Yin Q, Wu L, Han L, Zheng X, Tong R, Li L, Bai L, Bian Y. Immune-related adverse events of immune checkpoint inhibitors: a review. Front Immunol 2023; 14:1167975. [PMID: 37304306 PMCID: PMC10247998 DOI: 10.3389/fimmu.2023.1167975] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Since the first Immune Checkpoint Inhibitor was developed, tumor immunotherapy has entered a new era, and the response rate and survival rate of many cancers have also been improved. Despite the success of immune checkpoint inhibitors, resistance limits the number of patients who can achieve a lasting response, and immune-related adverse events complicate treatment. The mechanism of immune-related adverse events (irAEs) is unclear. We summarize and discuss the mechanisms of action of immune checkpoint inhibitors, the different types of immune-related adverse events and their possible mechanisms, and describe possible strategies and targets for prevention and therapeutic interventions to mitigate them.
Collapse
Affiliation(s)
- Qinan Yin
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liuyun Wu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lizhu Han
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xingyue Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lian Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Bai
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
57
|
Chen P, Li J, Tan H. Progress and Challenges of Immune Checkpoint Inhibitor-Induced Hypophysitis. J Clin Med 2023; 12:jcm12103468. [PMID: 37240574 DOI: 10.3390/jcm12103468] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/13/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a new type of antitumor drug which can achieve antitumor goals by blocking the binding of immune checkpoints to their ligands, thereby enhancing the activity of T cells. Meanwhile, ICIs block the binding of immune checkpoints to their ligands, disrupting the immune tolerance of T cells to self-antigens, which may lead to a series of immune-related adverse events (irAEs). Immune checkpoint inhibitor-induced hypophysitis (IH) is a relatively rare irAE. Due to the lack of specificity in clinical manifestations, it is difficult to accurately diagnose IH in a timely manner in clinical practice. However, the risk of adverse events, especially IH, for patients receiving ICIs has not been adequately investigated. Missed or delayed diagnosis may lead to poor prognosis or even adverse clinical outcomes. In this article, we summarize the epidemiology, pathogenesis, clinical manifestations, diagnosis and treatment of IH.
Collapse
Affiliation(s)
- Piaohong Chen
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianwei Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Pituitary Adenomas and Related Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huiwen Tan
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Pituitary Adenomas and Related Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
58
|
Uher O, Hadrava Vanova K, Lencova R, Frejlachova A, Wang H, Zhuang Z, Zenka J, Pacak K. Intratumoral immunotherapy of murine pheochromocytoma shows no age-dependent differences in its efficacy. Front Endocrinol (Lausanne) 2023; 14:1030412. [PMID: 37342258 PMCID: PMC10277857 DOI: 10.3389/fendo.2023.1030412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 04/18/2023] [Indexed: 06/22/2023] Open
Abstract
Cancer immunotherapy has shown remarkable clinical progress in recent years. Although age is one of the biggest leading risk factors for cancer development and older adults represent a majority of cancer patients, only a few new cancer immunotherapeutic interventions have been preclinically tested in aged animals. Thus, the lack of preclinical studies focused on age-dependent effect during cancer immunotherapy could lead to different therapeutic outcomes in young and aged animals and future modifications of human clinical trials. Here, we compare the efficacy of previously developed and tested intratumoral immunotherapy, based on the combination of polysaccharide mannan, toll-like receptor ligands, and anti-CD40 antibody (MBTA immunotherapy), in young (6 weeks) and aged (71 weeks) mice bearing experimental pheochromocytoma (PHEO). The presented results point out that despite faster growth of PHEO in aged mice MBTA intratumoral immunotherapy is effective approach without age dependence and could be one of the possible therapeutic interventions to enhance immune response to pheochromocytoma and perhaps other tumor types in aged and young hosts.
Collapse
Affiliation(s)
- Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Katerina Hadrava Vanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Radka Lencova
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Andrea Frejlachova
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Herui Wang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jan Zenka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
59
|
Bessone F, Björnsson ES. Drug-Induced Liver Injury due to Biologics and Immune Check Point Inhibitors. Med Clin North Am 2023; 107:623-640. [PMID: 37001957 DOI: 10.1016/j.mcna.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Biological agents have in the last two decades become very important therapeutic agents, particularly for the treatment of various autoimmune disorders. The most widely used biologics are the tumor necrosis factor-α (TNF-α) receptor antagonists: infliximab, adalimumab, and etanercept. Other commonly used biological agents are interleukin (IL)-1 receptor antagonist (Anakinra), interleukin (IL)-6 receptor antagonist (tocilizumab), and CD20 surface antigen antagonist (rituximab). The current review will however focus on TNF-α receptor antagonists.
Collapse
Affiliation(s)
- Fernando Bessone
- Facultad de Ciencias Médicas, Hospital Provincial del Centenario, University of Rosario, School of Medicine, Urquiza 3101, Rosario 2000, Argentina.
| | - Einar S Björnsson
- University of Iceland, Hringbraut 101, Reykjavik, Iceland; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landspitali University Hospital, Reykjavik, Iceland
| |
Collapse
|
60
|
Ascierto PA, Blank C, Eggermont AM, Garbe C, Gershenwald JE, Hamid O, Hauschild A, Luke JJ, Mehnert JM, Sosman JA, Tawbi HA, Mandalà M, Testori A, Caracò C, Osman I, Puzanov I. The "Great Debate" at Melanoma Bridge 2022, Naples, December 1st-3rd, 2022. J Transl Med 2023; 21:265. [PMID: 37072748 PMCID: PMC10114457 DOI: 10.1186/s12967-023-04100-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/30/2023] [Indexed: 04/20/2023] Open
Abstract
The Great Debate session at the 2022 Melanoma Bridge congress (December 1-3) featured counterpoint views from leading experts on five contemporary topics of debate in the management of melanoma. The debates considered the choice of anti-lymphocyte-activation gene (LAG)-3 therapy or ipilimumab in combination with anti-programmed death (PD)-1 therapy, whether anti-PD-1 monotherapy is still acceptable as a comparator arm in clinical trials, whether adjuvant treatment of melanoma is still a useful treatment option, the role of adjuvant therapy in stage II melanoma, what role surgery will continue to have in the treatment of melanoma. As is customary in the Melanoma Bridge Great Debates, the speakers are invited by the meeting Chairs to express one side of the assigned debate and the opinions given may not fully reflect personal views. Audiences voted in favour of either side of the argument both before and after each debate.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy.
| | | | - Alexander M Eggermont
- University Medical Center Utrecht & Princess Maxima Center, Utrecht, The Netherlands
- Comprehensive Cancer Center München, Technical University München & Ludwig Maximiliaan University, München, Germany
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omid Hamid
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, CA, USA
| | - Axel Hauschild
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Jason J Luke
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, USA
| | - Janice M Mehnert
- Perlmutter Cancer Center of NYU Langone/NYU Grossman School of Medicine, New York, NY, USA
| | - Jeffrey A Sosman
- Robert H Lurie Comprehensive Cancer Center, Northwestern University Medical Center, Chicago, IL, USA
| | - Hussein A Tawbi
- MD Anderson Brain Metastasis Clinic UT, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Alessandro Testori
- Image regenerative clinic Milan, Italy; EORTC Melanoma Group, Brussels, Belgium
| | - Corrado Caracò
- Division of Surgery of Melanoma and Skin Cancer, Istituto Nazionale Tumori "Fondazione Pascale" IRCCS, Naples, Italy
| | - Iman Osman
- Rudolf L. Baer, NYU Langone Medical Center, New York, NY, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
61
|
Alimonti P, Gonzalez Castro LN. The Current Landscape of Immune Checkpoint Inhibitor Immunotherapy for Primary and Metastatic Brain Tumors. Antibodies (Basel) 2023; 12:antib12020027. [PMID: 37092448 PMCID: PMC10123751 DOI: 10.3390/antib12020027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/14/2023] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Antibodies against immune checkpoint inhibitors (ICIs) have revolutionized the treatment of multiple aggressive malignancies, including melanoma and non-small cell lung cancer. ICIs for the treatment of primary and metastatic brain tumors have been used with varying degrees of success. Here, we discuss the available evidence for the use of ICIs in the treatment of primary and metastatic brain tumors, highlighting challenges and opportunities for furthering this type of cancer immunotherapy in neuro-oncology.
Collapse
Affiliation(s)
- Paolo Alimonti
- Department of Medicine, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milano, Italy
| | - L Nicolas Gonzalez Castro
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
62
|
Lai-Kwon J, Jacques S, Carlino M, Benannoune N, Robert C, Allayous C, Baroudjian B, Lebbe C, Zimmer L, Eroglu Z, Topcu TO, Dimitriou F, Haydon A, Lo SN, Menzies AM, Long GV. Efficacy of ipilimumab 3mg/kg following progression on low dose ipilimumab in metastatic melanoma. Eur J Cancer 2023; 186:12-21. [PMID: 37018924 DOI: 10.1016/j.ejca.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/15/2023] [Accepted: 03/04/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Differing doses of ipilimumab (IPI) are used in combination with an anti-PD1 antibody in advanced melanoma. There is no data on the outcomes of patients who progress following low-dose IPI (< 3 mg/kg) and are subsequently treated with IPI 3 mg/kg (IPI3). We conducted a multicentre retrospective survey to assess the efficacy of this strategy. METHODS Patients with resected stage III, unresectable stage III or IV melanoma who received low dose IPI (< 3 mg/kg) with an anti-PD1 antibody with recurrence (neo/adjuvant) or progressive disease (metastatic), who then received IPI3± anti-PD1 antibody were eligible. Best investigator-determined Response Evaluation Criteria in Solid Tumours response, progression-free survival (PFS) and overall survival (OS) were analysed. RESULTS Total 36 patients received low-dose IPI with an anti-PD1 antibody, 18 (50%) in the neo/adjuvant and 18 (50%) in the metastatic setting. Of which, 20 (56%) had primary resistance and 16 (44%) had acquired resistance. All patients received IPI3 for unresectable stage III or IV melanoma; median age 60 (29-78), 18 (50%) M1d disease, 32 (89%) Eastern Cooperative Oncology Group performance status 0-1. Around 35 (97%) received IPI3 with nivolumab and 1 received IPI3 alone. The response rate to IPI3 was 9/36 (25%). In patients with primary resistance, the response rate was 6/20 (30%). After a median follow-up of 22 months (95% CI: 15-27 months), the median PFS and OS were not reached in patients who responded; 1-year PFS and OS were 73% and 100%, respectively. CONCLUSIONS IPI3 following recurrence/progression on low dose IPI has clinical activity, including in primary resistance. IPI dosing is therefore critical in a subset of patients.
Collapse
|
63
|
Egeler MD, van Leeuwen M, Fraterman I, van den Heuvel NMJ, Boekhout AH, Lai-Kwon J, Wilthagen EA, Eriksson H, Haanen JB, Wilgenhof S, Ascierto PA, van Akkooi ACJ, van de Poll-Franse LV. Common toxicities associated with immune checkpoint inhibitors and targeted therapy in the treatment of melanoma: A systematic scoping review. Crit Rev Oncol Hematol 2023; 183:103919. [PMID: 36736511 DOI: 10.1016/j.critrevonc.2023.103919] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/23/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION This systematic scoping review compares the toxicities experienced by patients receiving immune checkpoint inhibitors (ICIs) or targeted therapy (TT) for stage III (resected and unresectable) and stage IV melanoma. METHODS OVID Medline, Embase, and PsycInfo were searched to identify Phase III trials reporting toxicities of FDA-approved ICIs and TT for advanced melanoma. AEs that were reported by ≥ 10% of patients in the evaluated trials were included. RESULTS Toxicity profiles of 11208 patients from 24 studies were reviewed. The rate of AEs was lower with ICIs compared to TT. However, ICIs were associated with higher rates of long-term or permanent AEs compared to TT, where toxicities generally were shortterm and reversible with treatment discontinuation. CONCLUSION The toxicity profiles of ICIs and TT vary substantially. Whilst the rate of AEs was lower with ICIs than during TT, it was also associated with higher rates of potentially chronic AEs.
Collapse
Affiliation(s)
- Mees D Egeler
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Marieke van Leeuwen
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Itske Fraterman
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Noelle M J van den Heuvel
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annelies H Boekhout
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Julia Lai-Kwon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Erica A Wilthagen
- Scientific Information Service, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hanna Eriksson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Medical Unit Head-Neck-, Lung-, Skin Cancer, Skin Cancer Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - John B Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Lonneke V van de Poll-Franse
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands; Department of Medical and Clinical Psychology, Center of Research on Psychology in Somatic diseases (CoRPS), Tilburg University, Tilburg, the Netherlands
| |
Collapse
|
64
|
Immune-related adverse events as potential surrogates of immune checkpoint inhibitors' efficacy: a systematic review and meta-analysis of randomized studies. ESMO Open 2023; 8:100787. [PMID: 36842300 PMCID: PMC9984799 DOI: 10.1016/j.esmoop.2023.100787] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/03/2022] [Accepted: 01/04/2023] [Indexed: 02/28/2023] Open
Abstract
BACKGROUND Immune-related adverse events (irAEs) are frequently reported during immune checkpoint inhibitor (ICI) therapy and are associated with long-term outcomes. It is unknown if the irAE occurrence is a valid surrogate of ICIs' efficacy. METHODS We identified articles reporting the results of randomized trials of experimental ICI therapy in solid tumors with a systematic search. The control arms could be placebo, cytotoxic/targeted therapy, or ICI therapy. We extracted the hazard ratios for overall survival (OS) with the number of OS events per arm and the number and percentages of overall and specific irAEs of grade 1-2 and grade 3-4 per arm. We estimated the treatment effect on the potential surrogate outcome with the odds ratio of the irAE rate between the experimental and the control arm. The statistical analysis consisted of weighted linear regression on a logarithmic scale between treatment effects on irAE rate and treatment effects on OS. RESULTS Sixty-two randomized trials were included for a total of 79 contrasts and 42 247 patients. The analyses found no significant association between the treatment effects for overall grade 1-2 or grade 3-4 irAE rates or specific (skin, gastrointestinal, endocrine) irAE rates. In the non-small-cell lung cancer (NSCLC) trial subset, we observed a negative association between treatment effects on overall grade 1-2 irAEs and treatment effects on OS in studies with patients selected for programmed death-ligand 1 expression (R2 = 0.55; 95% confidence interval 0.20-0.95; R = -0.69). In the melanoma trial subset, a negative association was shown between treatment effects on gastrointestinal grade 3-4 irAEs and treatment effects on OS in trials without an ICI-based control arm (R2 = 0.77; 95% confidence interval 0.24-0.99; R = -0.89). CONCLUSIONS We found low-strength correlations between the ICI therapy effects on overall or specific irAE rates and the treatment effects on OS in several cancer types.
Collapse
|
65
|
Knight A, Karapetyan L, Kirkwood JM. Immunotherapy in Melanoma: Recent Advances and Future Directions. Cancers (Basel) 2023; 15:1106. [PMID: 36831449 PMCID: PMC9954703 DOI: 10.3390/cancers15041106] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The use of immunotherapy in the treatment of advanced and high-risk melanoma has led to a striking improvement in outcomes. Although the incidence of melanoma has continued to rise, median survival has improved from approximately 6 months to nearly 6 years for patients with advanced inoperable stage IV disease. Recent understanding of the tumor microenvironment and its interplay with the immune system has led to the explosive development of novel immunotherapy treatments. Since the approval of the therapeutic cytokines interleukin-2 and interferon alfa-2 in the 1990s, the development of novel immune checkpoint inhibitors (ICIs), oncolytic virus therapy, and modulators of the tumor microenvironment have given way to a new era in melanoma treatment. Monoclonal antibodies directed at programmed cell death protein 1 receptor (PD-1) and its ligand (PDL-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and lymphocyte-activation gene 3 (LAG-3) have provided robust activation of the adaptive immune system, restoring immune surveillance leading to host tumor recognition and destruction. Multiple other immunomodulatory therapeutics are under investigation to overcome resistance to ICI therapy, including the toll-like receptor-9 (TLR-9) and 7/8 (TLR-7/8) agonists, stimulator of interferon genes (STING) agonists, and fecal microbiota transplantation. In this review, we focus on the recent advances in immunotherapy for the treatment of melanoma and provide an update on novel therapies currently under investigation.
Collapse
Affiliation(s)
- Andrew Knight
- Department of Medicine, Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Lilit Karapetyan
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M. Kirkwood
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
66
|
Li Y, Liang X, Li H, Chen X. Comparative efficacy and safety of immune checkpoint inhibitors for unresectable advanced melanoma: A systematic review and network meta-analysis. Int Immunopharmacol 2023; 115:109657. [PMID: 36608446 DOI: 10.1016/j.intimp.2022.109657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have entered the treatment paradigm for unresectable advanced melanoma, but there is a lack of evidence regarding its relative efficacy and safety. This study aim to compare the efficacy and safety of ICIs in patients with advanced unresectable melanoma. METHODS Studies included randomized clinical trials (RCTs) that compared ICIs, or combination therapy of ICIs, or with chemotherapy drugs, different ICIs, or one of the ICIs at different dosing schedules. Random-effects models of Bayesian network meta-analysis were performed following the PRISMA reporting guideline. Primary outcomes were overall survival (OS) and progression-free survival (PFS). Secondary outcomes included objective response rate (ORR), disease control rate (DCR), and adverse events. PROSPERO CRD42021229086. RESULTS Twenty-four RCTs with 18 different treatment regimens for advanced melanoma involving 10,090 patients were included. Overall, nivolumab 1 mg/kg plus ipilimumab 3 mg/kg treatment regimen were associated with the highest beneficial effect on OS, PFS, and DCR. Closely followed by nivolumab 3 mg/kg plus ipilimumab 1 mg/kg, and nivolumab plus relatlimab treatment regimens. However, three regimens had less favorable safety profiles. Although ipilimumab 0.3 mg/kg was ranked as the best options with the lowest risk of grade ≥ 3 treatment or immune-related adverse events, less therapeutic benefit was performed. The pembrolizumab 10 mg/kg regimen may be the preferred treatment with relative higher efficiency and safety among the ICIs regimens reported, as well as the nivolumab 3 mg/kg regimen. Head-to-head trials showed similar results. CONCLUSIONS This study shown the preferred treatment regimens with relatively higher efficiency and safety among the reported ICI regimens. Our results may complement the current standard of care, while its direct drug comparisons will aid future trials.
Collapse
Affiliation(s)
- Yan Li
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Xueyan Liang
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Huijuan Li
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Xiaoyu Chen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China; Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
67
|
Soltantabar P, Lon HK, Parivar K, Wang DD, Elmeliegy M. Optimizing benefit/risk in oncology: Review of post-marketing dose optimization and reflections on the road ahead. Crit Rev Oncol Hematol 2023; 182:103913. [PMID: 36681205 DOI: 10.1016/j.critrevonc.2023.103913] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Oncology therapies shifted from chemotherapy to molecularly targeted agents and finally to the era of immune-oncology agents. In contrast to cytotoxic agents, molecularly targeted agents are more selective, exhibit a wider therapeutic window, and may maximally modulate tumor growth at doses lower than the maximum tolerated dose (MTD). However, first-in-patient oncology studies for molecularly targeted agents continued to evaluate escalating doses using limited number of patients per dose cohort assessing dose-limiting toxicities to identify the MTD which is commonly selected for further development adopting a 'more is better' approach that led to several post-marketing requirement (PMR) studies to evaluate alternative, typically lower, doses or dosing frequencies to optimize the benefit-risk profile. In this review, post-marketing dose optimization efforts were reviewed including those required by a regulatory pathway or voluntarily conducted by the sponsor to improve efficacy, safety, or method of administration. Lessons learned and future implications from this deep dive review are discussed considering the evolving regulatory landscape on dose optimization for oncology compounds.
Collapse
Affiliation(s)
| | - Hoi-Kei Lon
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | | - Diane D Wang
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | |
Collapse
|
68
|
Williams KC, Gault A, Anderson AE, Stewart CJ, Lamb CA, Speight RA, Rajan N, Plummer R, Pratt AG. Immune-related adverse events in checkpoint blockade: Observations from human tissue and therapeutic considerations. Front Immunol 2023; 14:1122430. [PMID: 36776862 PMCID: PMC9909476 DOI: 10.3389/fimmu.2023.1122430] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Checkpoint inhibitors (CPIs) are monoclonal antibodies which, by disrupting interactions of immune checkpoint molecules with their ligands, block regulatory immune signals otherwise exploited by cancers. Despite revolutionary clinical benefits, CPI use is associated with an array of immune-related adverse events (irAEs) that mirror spontaneous autoreactivity. Severe irAEs necessitate pausing or stopping of CPI therapy and use of corticosteroids and/or other immunomodulatory interventions. Despite increasingly widespread CPI use, irAE pathobiology remains poorly understood; its elucidation may point to targeted mitigation strategies and uncover predictive biomarkers for irAE onset in patients, whilst casting new light on mechanisms of spontaneous immune-mediated disease. This review focuses on common CPI-induced irAEs of the gut, skin and synovial joints, and how these compare to immune-mediated diseases such as ulcerative colitis, vitiligo and inflammatory arthritis. We review current understanding of the immunological changes reported following CPI therapy at the level of peripheral blood and tissue. Many studies highlight dysregulation of cytokines in irAE-affected tissue, particularly IFNγ and TNF. IrAE-affected tissues are also predominantly infiltrated by T-cells, with low B-cell infiltration. Whilst there is variability between studies, patients treated with anti-programmed cell death-1 (PD-1)/PDL-1 therapies seem to exhibit CD8+ T-cell dominance, with CD4+ T-cells dominating in those treated with anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) monotherapy. Interestingly, CD8+CXCR3+ T-cells have been reported to be elevated in gastrointestinal, dermatological and musculoskeletal -irAE affected tissues. These findings may highlight potential opportunities for therapeutic development or re-deployment of existing therapies to prevent and/or improve the outcome of irAEs.
Collapse
Affiliation(s)
- Kristian C. Williams
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Abigail Gault
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Amy E. Anderson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher J. Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher A. Lamb
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - R. Ally Speight
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Neil Rajan
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Dermatology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Ruth Plummer
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Arthur G. Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Directorate of Musculoskeletal Services, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
69
|
Meng L, Wei Y, Xiao Y. Chemo-immunoablation of solid tumors: A new concept in tumor ablation. Front Immunol 2023; 13:1057535. [PMID: 36713427 PMCID: PMC9878389 DOI: 10.3389/fimmu.2022.1057535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
Chemical ablation was designed to inject chemical agents directly into solid tumors to kill cells and is currently only used clinically for the palliative treatment of tumors. The application and combination of different drugs, from anhydrous ethanol, and glacial acetic acid to epi-amycin, have been clinically tested for a long time. The effectiveness is unsatisfactory due to chemical agents' poor diffusion and concentration. Immunotherapy is considered a prospective oncologic therapeutic. Still, the clinical applications were limited by the low response rate of patients to immune drugs and the immune-related adverse effects caused by high doses. The advent of intratumoral immunotherapy has well addressed these issues. However, the efficacy of intratumoral immunotherapy alone is uncertain, as suggested by the results of preclinical and clinical studies. In this study, we will focus on the research of immunosuppressive tumor microenvironment with chemoablation and intratumoral immunotherapy, the synergistic effect between chemotherapeutic drugs and immunotherapy. We propose a new concept of intratumoral chemo-immunoablation. The concept opens a new perspective for tumor treatment from direct killing of tumor cells while, enhancing systemic anti-tumor immune response, and significantly reducing adverse effects of drugs.
Collapse
Affiliation(s)
- Liangliang Meng
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Radiology, Chinese PAP Hospital of Beijing, Beijing, China
| | - Yingtian Wei
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yueyong Xiao
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,*Correspondence: Yueyong Xiao,
| |
Collapse
|
70
|
Liu Z, Zhu Y, Xie H, Zou Z. Immune-mediated hepatitis induced by immune checkpoint inhibitors: Current updates and future perspectives. Front Pharmacol 2023; 13:1077468. [PMID: 36699050 PMCID: PMC9868416 DOI: 10.3389/fphar.2022.1077468] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
In recent years, cancer immunotherapy has made remarkable achievements. Immune checkpoint inhibitors (ICIs) have been used successfully in several types of cancer in the past decade. However, expanded indication and increased use of Immune checkpoint inhibitors have resulted in increased reports of toxicity called immune-related adverse events (irAEs). Due to the unique immunological characteristics of the liver, a hepatic immune-related adverse events has also been reported, which is usually termed Immune-mediated hepatitis (IMH). So far, it is generally considered that the mechanism of IMH induced by Immune checkpoint inhibitors is mainly the overactivation of T cells. It has been reported that the incidence of IMH ranges from 1% to 15%. Because of the lack of specific markers, a diagnosis of exclusion of IMH is critical. Although most IMH is mild and recoverable, several death cases have been reported, which has been increasingly concerned. This review summarizes the current understanding of the pathophysiology, epidemiology, diagnosis, management and prognosis of IMH caused by Immune checkpoint inhibitors. It also discusses the controversial issues in IMH, such as the role of liver biopsy, grading criteria, risk factors, rational treatment strategies with steroids, and the timing of Immune checkpoint inhibitors rechallenging, which may provide helpful information for IMH in future clinical practice.
Collapse
Affiliation(s)
- Zherui Liu
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China
| | - Yun Zhu
- Medical School of Chinese PLA, Beijing, China
| | - Huan Xie
- Medical School of Chinese PLA, Beijing, China
| | - Zhengsheng Zou
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China,*Correspondence: Zhengsheng Zou,
| |
Collapse
|
71
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
72
|
Vidovic D, Helyer LK, Pasternak S, Giacomantonio CA. Abscopal responses in patients with metastatic melanoma involving skin and subcutaneous tissues treated with intralesional IL2 plus BCG. Front Oncol 2023; 13:1160269. [PMID: 37182189 PMCID: PMC10172468 DOI: 10.3389/fonc.2023.1160269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Cutaneous melanoma is relatively common with increasing incidence and significant mortality. While the mainstay of therapy is surgical, patients with stage III and IV disease fare poorer than those with early-stage disease and often benefit from adjuvant therapies. While systemic immunotherapy has changed the landscape of melanoma treatment, for some patients systemic toxicities related to these treatments prohibit successful administration or completion of therapy. Moreover, it is becoming increasingly evident that nodal, regional, and in-transit disease appears to be resistant to systemic immunotherapy relative to responses observed in distant metastatic disease sites. In this scenario, intralesional immunotherapies may offer benefit. In this case series, we describe the use of intralesional IL-2 and BCG at our institution in ten patients with in-transit plus or minus distant cutaneous metastatic melanoma over the last twelve years. All patients received intralesional IL2 and BCG. Both treatments were very well tolerated with only grade 1/2 adverse events. In our cohort, complete clinical response was 60% (6/10), progressive disease in 20% (2/10), and no response in 20% (2/10) of patients. The overall response rate (ORR) was 70%. The median overall survival was 35.5 months and mean overall survival 43 months in this cohort. Herein we further highlight the clinical, histopathological, and radiological course of two complete responders, showing evidence of an abscopal effect with resolution of distant untreated metastasis. Together, this limited data supports the safe and effective use of intralesional IL2 and BCG for the treatment of metastatic or in-transit melanoma in this challenging patient cohort. To our knowledge, this is the first formal study to report on this combination therapy for the treatment of melanoma.
Collapse
Affiliation(s)
- Dejan Vidovic
- Division of General and Gastrointestinal Surgery, Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Lucy Kathryn Helyer
- Division of General and Gastrointestinal Surgery, Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Sylvia Pasternak
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Carman Anthony Giacomantonio
- Division of General and Gastrointestinal Surgery, Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- *Correspondence: Carman Anthony Giacomantonio,
| |
Collapse
|
73
|
Jacques JP, Valadares LP, Moura AC, Oliveira MRF, Naves LA. Frequency and clinical characteristics of hypophysitis and hypopituitarism in patients undergoing immunotherapy - A systematic review. Front Endocrinol (Lausanne) 2023; 14:1091185. [PMID: 36875457 PMCID: PMC9974646 DOI: 10.3389/fendo.2023.1091185] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023] Open
Abstract
OBJECTIVE To describe the frequency of hypophysitis and hypopituitarism in cancer patients who are undergoing antineoplastic treatment with immunotherapy, as well as to describe the clinical, epidemiological, and demographic characteristics of these patients. METHODS A systematic search of the literature in PubMed, Embase, Web of Science, ClinicalTrials.gov and Cochrane Controlled Register of Trials took place on May 8 and 9, 2020. Randomized and nonrandomized clinical trials, cohort studies, case-control studies, case series and case reports were included. RESULTS A total of 239 articles were obtained, in which 963 cases of hypophysitis and 128 cases of hypopituitarism were found in a treated population of 30,014 individuals (3.20% and 0.42% of the evaluated population, respectively). In the cohort studies, the incidence of hypophysitis and hypopituitarism ranged from 0% to 27.59% and from 0% to 17.86%, respectively. In the non-randomized clinical trials, the incidence of hypophysitis and hypopituitarism ranged from 0% to 25% and from 0% to 14.67%, and in randomized clinical trials from 0% to 16.2% and from 0% to 33.33%. The most common hormonal changes were in the corticotrophic, thyrotrophic and gonadotrophic axes. The main magnetic resonance imaging (MRI) findings were enlargement of the pituitary gland and enhanced contrast uptake. The main symptoms presented by patients with hypophysitis were fatigue and headache. CONCLUSION The present review reported a frequency of hypophysitis and hypopituitarism of 3.20% and 0.42%, respectively, in the evaluated population. The clinical-epidemiological characteristics of patients affected by hypophysitis were also described. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/, identifier CRD42020175864.
Collapse
Affiliation(s)
- Juliana Prudêncio Jacques
- Faculty of Health Science, University of Brasilia, Brasilia, Brazil
- *Correspondence: Juliana Prudêncio Jacques,
| | - Luciana Pinto Valadares
- Department of Internal Medicine and Endocrinology, Sarah Network of Rehabilitation Hospitals, Brasília, Brazil
| | | | | | | |
Collapse
|
74
|
Plunkett KR, Armitage JD, Inderjeeth AJ, McDonnell AM, Waithman J, Lau PKH. Tissue-resident memory T cells in the era of (Neo) adjuvant melanoma management. Front Immunol 2022; 13:1048758. [PMID: 36466880 PMCID: PMC9709277 DOI: 10.3389/fimmu.2022.1048758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/13/2022] [Indexed: 10/10/2023] Open
Abstract
Tissue-resident memory T (TRM) cells have emerged as key players in the immune control of melanoma. These specialized cells are identified by expression of tissue retention markers such as CD69, CD103 and CD49a with downregulation of egress molecules such as Sphingosine-1-Phosphate Receptor-1 (S1PR1) and the lymphoid homing receptor, CD62L. TRM have been shown to be integral in controlling infections such as herpes simplex virus (HSV), lymphocytic choriomeningitis virus (LCMV) and influenza. More recently, robust pre-clinical models have also demonstrated TRM are able to maintain melanoma in a dormant state without progression to macroscopic disease reminiscent of their ability to control viral infections. The discovery of the role these cells play in anti-melanoma immunity has coincided with the advent of immune checkpoint inhibitor (ICI) therapy which has revolutionized the treatment of cancers. ICIs that target programmed death protein-1 (PD-1) and cytotoxic T lymphocyte antigen-4 (CTLA-4) have led to substantial improvements in outcomes for patients with metastatic melanoma and have been rapidly employed to reduce recurrences in the resected stage III setting. While ICIs mediate anti-tumor activity via CD8+ T cells, the specific subsets that facilitate this response is unclear. TRM invariably exhibit high expression of immune checkpoints such as PD-1, CTLA-4 and lymphocyte activating gene-3 (LAG-3) which strongly implicates this CD8+ T cell subset as a crucial mediator of ICI activity. In this review, we present pre-clinical and translational studies that highlight the critical role of TRM in both immune control of primary melanoma and as a key CD8+ T cell subset that mediates anti-tumor activity of ICIs for the treatment of melanoma.
Collapse
Affiliation(s)
- Kai R. Plunkett
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Jesse D. Armitage
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | | | - Alison M. McDonnell
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Jason Waithman
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Peter K. H. Lau
- Melanoma Discovery Laboratory, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| |
Collapse
|
75
|
Early Effects of Nivolumab and Ipilimumab Combined Immunotherapy in the Treatment of Metastatic Melanoma in Poland: A Multicenter Experience. Biomedicines 2022; 10:biomedicines10102528. [PMID: 36289790 PMCID: PMC9598919 DOI: 10.3390/biomedicines10102528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Nivolumab and ipilimumab combination became the first-line standard in advanced melanoma. We assessed its efficacy in a real-life study in Poland. In a one-year follow-up, we evaluated the medical records of 50 melanoma patients treated with that modality in five oncology centers. We recorded therapy outcomes and adverse events (AEs) after 3 and 12 months of therapy. At the first checkpoint, the disease control rate (DCR) was recorded in 58% (n = 29) of patients, but the same number of patients (n = 29, 58%) stopped immunotherapy due to disease progression (PD, n = 14, 48.3%), toxicity (n = 11, 37.9%) or death (n = 4, 13.8%). Among patients with DCR after the induction phase, 8 (27.6%) terminated due to toxicity, and 21 (72.4%) continued. However, at the 12-month checkpoint, only 14 patients (27% of all) were still receiving immunotherapy. In 7 (33.3%) it was discontinued due to PD (n = 2), toxicity (n = 2, 28.6% each), or death (n = 3, 42.9%). AEs occurred in 66.7% (n = 34) of patients; severe (grade 3 or 4) in half of them. Interestingly, those with AEs had an 80% lower risk of death (hazard ratio [HR] 0.2, 95% confidence interval [CI] 0.07−0.57, p = 0.001) and PD (HR 0.2, 95%CI 0.09−0.47, p < 0.0001). In the entire group of patients, after a 12-month follow-up, the median overall survival was not reached (NR, range: 6.8 months-NR) and progression-free survival was 6.3 (range: 3-NR) months. Our results demonstrate that combined immunotherapy is less effective in real-life than in pivotal trials. However, early responders will likely continue the therapy after a one-year follow-up. AEs occurrence might be a predictor of clinical effectiveness.
Collapse
|
76
|
Hauguel C, Ducellier S, Provot O, Ibrahim N, Lamaa D, Balcerowiak C, Letribot B, Nascimento M, Blanchard V, Askenatzis L, Levaique H, Bignon J, Baschieri F, Bauvais C, Bollot G, Renko D, Deroussent A, Prost B, Laisne MC, Michallet S, Lafanechère L, Papot S, Montagnac G, Tran C, Alami M, Apcher S, Hamze A. Design, synthesis and biological evaluation of quinoline-2-carbonitrile-based hydroxamic acids as dual tubulin polymerization and histone deacetylases inhibitors. Eur J Med Chem 2022; 240:114573. [DOI: 10.1016/j.ejmech.2022.114573] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/04/2022]
|
77
|
Yu L, Sun M, Zhang Q, Zhou Q, Wang Y. Harnessing the immune system by targeting immune checkpoints: Providing new hope for Oncotherapy. Front Immunol 2022; 13:982026. [PMID: 36159789 PMCID: PMC9498063 DOI: 10.3389/fimmu.2022.982026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
With the goal of harnessing the host's immune system to provide long-lasting remission and cures for various cancers, the advent of immunotherapy revolutionized the cancer therapy field. Among the current immunotherapeutic strategies, immune checkpoint blockades have greatly improved the overall survival rates in certain patient populations. Of note, CTLA4 and PD-1/PD-L1 are two major non-redundant immune checkpoints implicated in promoting cancer immune evasion, and ultimately lead to relapse. Antibodies or inhibitors targeting these two c+heckpoints have achieved some encouraging clinical outcomes. Further, beyond the canonical immune checkpoints, more inhibitory checkpoints have been identified. Herein, we will summarize recent progress in immune checkpoint blockade therapies, with a specific focus on key pre-clinical and clinical results of new immune checkpoint therapies for cancer. Given the crucial roles of immune checkpoint blockade in oncotherapy, drugs targeting checkpoint molecules expressed by both cancer and immune cells are in clinical trials, which will be comprehensively summarized in this review. Taken together, investigating combinatorial therapies targeting immune checkpoints expressed by cancer cells and immune cells will greatly improve immunotherapies that enhance host elimination of tumors.
Collapse
Affiliation(s)
- Lu Yu
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Minghan Sun
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Institute of Organ Transplantation, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
78
|
van Not OJ, Blokx WAM, van den Eertwegh AJM, de Meza MM, Haanen JB, Blank CU, Aarts MJB, van den Berkmortel FWPJ, de Groot JWB, Hospers GAP, Kapiteijn E, Piersma D, van Rijn RS, Stevense-den Boer M, van der Veldt AAM, Boers-Sonderen MJ, Jansen AML, Wouters MWJM, Suijkerbuijk KPM. BRAF and NRAS Mutation Status and Response to Checkpoint Inhibition in Advanced Melanoma. JCO Precis Oncol 2022; 6:e2200018. [PMID: 36130145 DOI: 10.1200/po.22.00018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Little is known about the effect of specific gene mutations on efficacy of immune checkpoint inhibitors in patients with advanced melanoma. MATERIALS AND METHODS All patients with advanced melanoma treated with first-line anti-PD-1 or ipilimumab-nivolumab between 2012 and 2021 in the nationwide Dutch Melanoma Treatment Registry were included in this cohort study. Objective response rate, progression-free survival (PFS), and overall survival (OS) were analyzed according to BRAF and NRAS status. A multivariable Cox model was used to analyze prognostic factors associated with PFS and OS. RESULTS In total, 1764 patients received anti-PD-1 and 759 received ipilimumab-nivolumab. No significant differences in PFS were found in the anti-PD-1 cohort. In the ipilimumab-nivolumab cohort, median PFS was significantly higher for BRAF-mutant melanoma (9.9 months; 95% CI, 6.8 to 17.2) compared with NRAS-mutant (4.8 months; 95% CI, 3.0 to 7.5) and double wild-type (5.3 months; 95% CI, 3.6 to 7.1). In multivariable analysis, BRAF-mutant melanoma was significantly associated with a lower risk of progression or death in the ipilimumab-nivolumab cohort. Median OS was significantly higher for BRAF-mutant melanoma compared with NRAS-mutant and double wild-type melanoma for both immune checkpoint inhibitor regimens. CONCLUSION Ipilimumab-nivolumab-treated patients with BRAF-mutant melanoma display improved PFS and OS compared with patients with NRAS-mutant and double wild-type melanoma. BRAF mutation status is a factor to consider while choosing between mono and dual checkpoint inhibition in advanced melanoma.
Collapse
Affiliation(s)
- Olivier J van Not
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, the Netherlands.,Department of Medical Oncology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Willeke A M Blokx
- Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Melissa M de Meza
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, the Netherlands.,Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands.,Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B Haanen
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christian U Blank
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Medical Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | | | | | - Geke A P Hospers
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Djura Piersma
- Department of Internal Medicine, Medisch Spectrum Twente, Enschede, the Netherlands
| | - Rozemarijn S van Rijn
- Department of Internal Medicine, Medical Centre Leeuwarden, Leeuwarden, the Netherlands
| | | | - Astrid A M van der Veldt
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Anne M L Jansen
- Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Michel W J M Wouters
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, the Netherlands.,Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands.,Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
79
|
Zhang R, Kong D, Chen R, Guo Y, Jian W, Han M, Zhou T. A model‐based meta‐analysis of immune‐related adverse events during immune checkpoint inhibitors treatment for
NSCLC. CPT Pharmacometrics Syst Pharmacol 2022; 11:1135-1146. [PMID: 35763678 PMCID: PMC9381889 DOI: 10.1002/psp4.12834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 11/07/2022] Open
Affiliation(s)
- Renwei Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Daming Kong
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Rong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Yuchen Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Weizhe Jian
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Mengyi Han
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Tianyan Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| |
Collapse
|
80
|
Immunotherapy-induced Colitis: A Comprehensive Review of Epidemiology, Clinical Presentation, Diagnostic Workup, and Management Plan. J Clin Gastroenterol 2022; 56:555-564. [PMID: 35470301 DOI: 10.1097/mcg.0000000000001705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of a variety of malignancies including advanced melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck cancers among others. Since their introduction, there has been significant improvement in survival and prognosis in patients with advanced malignancies. Unfortunately, improved outcomes have come at a price of significant immune-related adverse events, with those of the gastrointestinal tract being the most common. Gastrointestinal immune-related adverse events frequently present as diarrhea and colitis, the severity of which can range from mild diarrhea to fulminant colitis with intestinal perforation. Currently, management of ICI-induced colitis is primarily guided by retrospective studies and expert opinion. A significant number of ICI-induced colitis responds to high-dose corticosteroids; however, some patients require further therapy with biologics. There is limited information on the factors which may predispose patients to ICI-induced colitis. Future research elucidating these risk factors along with development of a scoring system could allow for risk-stratification of patients before initiation of ICI therapy. Such a system may help clinicians and patients keep a high index of suspicion regarding ICI-induced colitis and could hopefully reduce the incidence of severe cases. Similarly, future studies should investigate protective factors against ICI-induced colitis, which could potentially allow more patients to safely benefit from ICI therapy.
Collapse
|
81
|
Chye A, Allen I, Barnet M, Burnett DL. Insights Into the Host Contribution of Endocrine Associated Immune-Related Adverse Events to Immune Checkpoint Inhibition Therapy. Front Oncol 2022; 12:894015. [PMID: 35912205 PMCID: PMC9329613 DOI: 10.3389/fonc.2022.894015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Blockade of immune checkpoints transformed the paradigm of systemic cancer therapy, enabling substitution of a cytotoxic chemotherapy backbone to one of immunostimulation in many settings. Invigorating host immune cells against tumor neo-antigens, however, can induce severe autoimmune toxicity which in many cases requires ongoing management. Many immune-related adverse events (irAEs) are clinically and pathologically indistinguishable from inborn errors of immunity arising from genetic polymorphisms of immune checkpoint genes, suggesting a possible shared driver for both conditions. Many endocrine irAEs, for example, have analogous primary genetic conditions with varied penetrance and severity despite consistent genetic change. This is akin to onset of irAEs in response to immune checkpoint inhibitors (ICIs), which vary in timing, severity and nature despite a consistent drug target. Host contribution to ICI response and irAEs, particularly those of endocrine origin, such as thyroiditis, hypophysitis, adrenalitis and diabetes mellitus, remains poorly defined. Improved understanding of host factors contributing to ICI outcomes is essential for tailoring care to an individual’s unique genetic predisposition to response and toxicity, and are discussed in detail in this review.
Collapse
Affiliation(s)
- Adrian Chye
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - India Allen
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
| | - Megan Barnet
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
- *Correspondence: Megan Barnet, ; Deborah L. Burnett,
| | - Deborah L. Burnett
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- *Correspondence: Megan Barnet, ; Deborah L. Burnett,
| |
Collapse
|
82
|
Issa M, Tang J, Guo Y, Coss C, Mace TA, Bischof J, Phelps M, Presley CJ, Owen DH. Risk factors and predictors of immune-related adverse events: implications for patients with non-small cell lung cancer. Expert Rev Anticancer Ther 2022; 22:861-874. [PMID: 35786142 DOI: 10.1080/14737140.2022.2094772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICI) are now utilized as a standard of care treatment for multiple cancers, including in both the metastatic setting as well as in earlier stages of disease. The identification of unique immune-related adverse events (irAE) that occur during ICI treatment has led to intense research to identify potential risk factors and biomarkers that may assist in clinical decision making. Although initial studies in ICI were primarily in advanced stage disease, the use of ICI in earlier stages of disease as adjuvant therapies requires a better understanding of patient risk stratification to mitigate or prevent serious irAE. AREAS COVERED In this review, we set out to describe the current state of research regarding potential risk factors for irAE in patients with non-small cell lung cancer, as well as explore the barriers to understanding irAE. We review data from irAE that occur in large phase 3 trials and prospective studies focusing on irAE, as well as the many retrospective studies that currently form the bulk of our understanding of irAE.
Collapse
Affiliation(s)
- Majd Issa
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Joy Tang
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Yizhen Guo
- College of Pharmacy, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Chris Coss
- College of Pharmacy, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Thomas A Mace
- Division of Gastroenterology, Hepatology & Nutrition, Department of Internal Medicine, the Ohio State University Wexner Medical Center, Columbus, USA
| | - Jason Bischof
- Department of Emergency Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Mitch Phelps
- College of Pharmacy, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Carolyn J Presley
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Dwight H Owen
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| |
Collapse
|
83
|
Holmberg CJ, Ny L, Hieken TJ, Block MS, Carr MJ, Sondak VK, Örtenwall C, Katsarelias D, Dimitriou F, Menzies AM, Saw RPM, Rogiers A, Straker RJ, Karakousis G, Applewaite R, Pallan L, Han D, Vetto JT, Gyorki DE, Tie EN, Vitale MG, Ascierto PA, Dummer R, Cohen J, Hui JYC, Schachter J, Asher N, Helgadottir H, Chai H, Kroon H, Coventry B, Rothermel LD, Sun J, Carlino MS, Duncan Z, Broman K, Weber J, Lee AY, Berman RS, Teras J, Ollila DW, Long GV, Zager JS, van Akkooi A, Olofsson Bagge R. The efficacy of immune checkpoint blockade for melanoma in-transit with or without nodal metastases - A multicenter cohort study. Eur J Cancer 2022; 169:210-222. [PMID: 35644725 PMCID: PMC9975793 DOI: 10.1016/j.ejca.2022.03.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/16/2022] [Accepted: 03/31/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Guidelines addressing melanoma in-transit metastasis (ITM) recommend immune checkpoint inhibitors (ICI) as a first-line treatment option, despite the fact that there are no efficacy data available from prospective trials for exclusively ITM disease. The study aims to analyze the outcome of patients with ITM treated with ICI based on data from a large cohort of patients treated at international referral clinics. METHODS A multicenter retrospective cohort study of patients treated between January 2015 and December 2020 from Australia, Europe, and the USA, evaluating treatment with ICI for ITM with or without nodal involvement (AJCC8 N1c, N2c, and N3c) and without distant disease (M0). Treatment was with PD-1 inhibitor (nivolumab or pembrolizumab) and/or CTLA-4 inhibitor (ipilimumab). The response was evaluated according to the RECIST criteria modified for cutaneous lesions. RESULTS A total of 287 patients from 21 institutions in eight countries were included. Immunotherapy was first-line treatment in 64 (22%) patients. PD-1 or CTLA-4 inhibitor monotherapy was given in 233 (81%) and 23 (8%) patients, respectively, while 31 (11%) received both in combination. The overall response rate was 56%, complete response (CR) rate was 36%, and progressive disease (PD) rate was 32%. Median PFS was ten months (95% CI 7.4-12.6 months) with a one-, two-, and five-year PFS rate of 48%, 33%, and 18%, respectively. Median MSS was not reached, and the one-, two-, and five-year MSS rates were 95%, 83%, and 71%, respectively. CONCLUSION Systemic immunotherapy is an effective treatment for melanoma ITM. Future studies should evaluate the role of systemic immunotherapy in the context of multimodality therapy, including locoregional treatments such as surgery, intralesional therapy, and regional therapies.
Collapse
Affiliation(s)
- Carl-Jacob Holmberg
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden,Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars Ny
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tina J. Hieken
- Department of Surgery, Mayo Clinic, Rochester, USA,Mayo Clinic Cancer Center, Rochester, USA
| | - Matthew S. Block
- Mayo Clinic Cancer Center, Rochester, USA,Department of Oncology, Mayo Clinic, Rochester, USA
| | - Michael J. Carr
- Department of Cutaneous Oncology Moffitt Cancer Center, Tampa, USA
| | - Vernon K. Sondak
- Department of Cutaneous Oncology Moffitt Cancer Center, Tampa, USA
| | - Christoffer Örtenwall
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Dimitrios Katsarelias
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Florentia Dimitriou
- Melanoma Institute Australia, The University of Sydney, Sydney Australia,Department of Dermatology, University Hospital of Zürich, Zürich, Switzerland
| | - Alexander M. Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia,Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Robyn PM. Saw
- Melanoma Institute Australia, The University of Sydney, Sydney Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Aljosja Rogiers
- Melanoma Institute Australia, The University of Sydney, Sydney Australia
| | - Richard J. Straker
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, USA
| | - Giorgos Karakousis
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, USA
| | - Rona Applewaite
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Lalit Pallan
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Dale Han
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, USA
| | - John T. Vetto
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, USA
| | - David E. Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department, University of Melbourne, Melbourne, Australia
| | - Emilia Nan Tie
- Division of Cancer Surgery, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department, University of Melbourne, Melbourne, Australia
| | - Maria Grazia Vitale
- Department of Skin Cancers, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Paulo A. Ascierto
- Department of Skin Cancers, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zürich, Zürich, Switzerland
| | - Jade Cohen
- Department of Surgery, University of Minnesota, Minneapolis, USA
| | - Jane YC. Hui
- Department of Surgery, University of Minnesota, Minneapolis, USA
| | - Jacob Schachter
- The Ella Lemelbaum Institite for Immuno-oncology, Sheba Medical Center, Tel Aviv, Israel
| | - Nethanel Asher
- The Ella Lemelbaum Institite for Immuno-oncology, Sheba Medical Center, Tel Aviv, Israel
| | - H. Helgadottir
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden,Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Harvey Chai
- Department of Surgery, Royal Adelaide Hospital, Adelaide, Australia,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Hidde Kroon
- Department of Surgery, Royal Adelaide Hospital, Adelaide, Australia,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Brendon Coventry
- Department of Surgery, Royal Adelaide Hospital, Adelaide, Australia,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Luke D. Rothermel
- Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, USA,Case Western Reserve University, Cleveland, USA
| | - James Sun
- Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, USA,Case Western Reserve University, Cleveland, USA
| | - Matteo S. Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney Australia,Department of Medical Oncology, Westmead and Blacktown Hospitals, The Crown Princess Mary Cancer Centre, Sydney, Australia
| | - Zoey Duncan
- University of Alabama at Birmingham, Birmingham, USA
| | - Kristy Broman
- University of Alabama at Birmingham, Birmingham, USA
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, USA
| | - Ann Y. Lee
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, USA,NYU Grossman School of Medicine, Department of Surgery, New York, USA
| | - Russell S. Berman
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, USA,NYU Grossman School of Medicine, Department of Surgery, New York, USA
| | - Jüri Teras
- North Estonian Medical Centre Foundation, Tallinn, Estonia
| | - David W. Ollila
- Division of Surgical Oncology and Endocrine Surgery, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Georgina V. Long
- Melanoma Institute Australia, The University of Sydney, Sydney Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia,Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Jonathan S. Zager
- Department of Cutaneous Oncology Moffitt Cancer Center, Tampa, USA,Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, USA
| | - Alexander van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
84
|
O'Quigley J. Testing for Differences in Survival When Treatment Effects Are Persistent, Decaying, or Delayed. J Clin Oncol 2022; 40:3537-3545. [PMID: 35767775 DOI: 10.1200/jco.21.01811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A statistical test for the presence of treatment effects on survival will be based on a null hypothesis (absence of effects) and an alternative (presence of effects). The null is very simply expressed. The most common alternative, also simply expressed, is that of proportional hazards. For this situation, not only do we have a very powerful test in the log-rank test but also the outcome is readily interpreted. However, many modern treatments fall outside this relatively straightforward paradigm and, as such, have attracted attention from statisticians eager to do their best to avoid losing power as well as to maintain interpretability when the alternative hypothesis is less simple. Examples include trials where the treatment effect decays with time, immunotherapy trials where treatment effects may be slow to manifest themselves as well as the so-called crossing hazards problem. We review some of the solutions that have been proposed to deal with these issues. We pay particular attention to the integrated log-rank test and how it can be combined with the log-rank test itself to obtain powerful tests for these more complex situations.
Collapse
Affiliation(s)
- John O'Quigley
- Department of Statistical Science, University College London, London, United Kingdom
| |
Collapse
|
85
|
Dong M, Yu T, Zhang Z, Zhang J, Wang R, Tse G, Liu T, Zhong L. ICIs-Related Cardiotoxicity in Different Types of Cancer. J Cardiovasc Dev Dis 2022; 9:203. [PMID: 35877565 PMCID: PMC9324462 DOI: 10.3390/jcdd9070203] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are rapidly developing immunotherapy cancer drugs that have prolonged patient survival. However, ICIs-related cardiotoxicity has been recognized as a rare, but fatal, consequence. Although there has been extensive research based on different types of ICIs, these studies have not indicated whether cardiotoxicity is specific to a type of cancer. Therefore, we conducted a systematic review to analyze a variety of ICIs-related cardiotoxicity, focusing on different types of cancer. We found that the incidence of ICIs-related cardiac adverse events (CAEs) and common cardiotoxic manifestations vary with cancer type. This inspired us to explore the underlying mechanisms to formulate targeted clinical strategies for maintaining the cardiovascular health of cancer patients.
Collapse
Affiliation(s)
- Mei Dong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| | - Ting Yu
- Medical College, Qingdao University, Qingdao 266003, China;
| | - Zhenzhen Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| | - Jing Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| | - Rujian Wang
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China;
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300070, China;
- Kent and Medway Medical School, Canterbury CT2 7FS, UK
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300070, China;
| | - Lin Zhong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| |
Collapse
|
86
|
Wankhede D, Grover S. Outcomes After Curative Metastasectomy for Patients with Malignant Melanoma: A Systematic Review and Meta-analysis. Ann Surg Oncol 2022; 29:3709-3723. [PMID: 35128602 DOI: 10.1245/s10434-022-11351-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2023]
Abstract
BACKGROUND Evidence on the role of curative metastasectomy (CM) for malignant melanoma (MM) patients is limited, especially in the current era of effective systemic therapy. A systematic review and meta-analysis were performed to ascertain the role of CM compared with incomplete or nonsurgical treatment for patients with MM. METHODS Medline, Embase, and Scopus databases were searched for studies investigating CM for MM until 30 September 2021. The review included studies that compared CM with no-CM and reported a hazard ratio (HR) after multivariate analysis for overall survival. A random-effects model with inverse variance was used to calculate pooled HR. The Newcastle-Ottawa Scale was used to assess the risk of bias. RESULTS For the final analysis, 40 studies including 31,282 patients (CM, 9958; no-CM, 21,324) were considered. Compared with no-CM, CM was associated with a significantly lower risk of death (HR, 0.42; 95% confidence interval [CI], 0.38-0.47; p < 0.00001). Subgroup analysis showed that the outcome was independent of the effective systemic therapy and anatomic location of metastasis. An unfavorable prognosis was associated with advancing age, elevated lactate dehydrogenase (LDH), male gender, prior stage 3 disease, multiple metastases and organ sites, and shorter disease-free interval. CONCLUSION Curative metastasectomy for MM is associated with a lower risk of death than non-curative treatment methods. Selection bias and underlying weakness of studies reduced the strength of evidence in this review. However, CM should be a part of the multimodality treatment of MM whenever technically feasible.
Collapse
Affiliation(s)
- Durgesh Wankhede
- Department of Surgical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India.
| | - Sandeep Grover
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
87
|
Yin J, Elias R, Peng L, Levonyak N, Asokan A, Christie A, Kubiliun N, Brugarolas J, Hammers HJ. Chronic Use of Proton Pump Inhibitors Is Associated With an Increased Risk of Immune Checkpoint Inhibitor Colitis in Renal Cell Carcinoma. Clin Genitourin Cancer 2022; 20:260-269. [PMID: 35277350 PMCID: PMC9701615 DOI: 10.1016/j.clgc.2022.01.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 12/20/2021] [Accepted: 01/25/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have become a standard of care in metastatic renal cell carcinoma (mRCC) but are associated with immune-related adverse events (irAEs) including colitis. Growing evidence suggests proton pump inhibitors (PPIs) increase the risk of inflammatory bowel disease (IBD). Given the pathophysiological overlap between IBD and ICI colitis, we sought to evaluate the relationship between PPI use and ICI colitis in mRCC patients. PATIENTS AND METHODS We performed a retrospective study of adult patients who received ICI therapy for mRCC between 2015 and 2018 at University of Texas Southwestern Medical Center affiliated hospitals. Clinical characteristics, oncological outcomes, ICI colitis details, and PPI use details were collected by manual chart review. The diagnosis of ICI colitis was made via biopsy when available, or by clinical criteria (symptoms and response to immunosuppressive therapy) when biopsy specimens were unavailable or inconclusive. Univariable and multivariable logistic regression analyses were conducted to assess the potential contribution of PPIs to ICI colitis. RESULTS A total of 176 patients received ICI therapy for mRCC, of which 16 (9.1%) were diagnosed with ICI colitis. Patients with ICI colitis presented with elevated stool lactoferritin and calprotectin and a wide range of endoscopic and histologic findings. There were no significant differences between patients with and without ICI colitis in age, gender, medical comorbidities, RCC history, and overall survival. However, exposure to ipilimumab and PPI use were more frequently observed in patients with ICI colitis than those without. In univariable and multivariable logistic regression analyses, exposure to ipilimumab and chronic use of PPIs > 8 weeks were significantly associated with ICI colitis. CONCLUSION In addition to ipilimumab use, chronic use of PPIs may be associated with ICI colitis in patients with mRCC.
Collapse
Affiliation(s)
- Jianyi Yin
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Roy Elias
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390,Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Lan Peng
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Nicholas Levonyak
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Annapoorani Asokan
- University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Alana Christie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Nisa Kubiliun
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390,Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Hans J Hammers
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX; Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
88
|
Iranzo P, Callejo A, Assaf JD, Molina G, Lopez DE, Garcia-Illescas D, Pardo N, Navarro A, Martinez-Marti A, Cedres S, Carbonell C, Frigola J, Amat R, Felip E. Overview of Checkpoint Inhibitors Mechanism of Action: Role of Immune-Related Adverse Events and Their Treatment on Progression of Underlying Cancer. Front Med (Lausanne) 2022; 9:875974. [PMID: 35707528 PMCID: PMC9189307 DOI: 10.3389/fmed.2022.875974] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
In recent years, immunotherapy-based regimens have been included into the treatment's algorithm of several cancer types. Programmed death-1 (PD-1) and cytotoxic T lymphocyte antigen-4 (CTLA-4) interact with their ligands found on the surface of antigen presenting cells (APC) or tumor cells (PD-L1/2 and CD80/86). Through these interactions, stimulatory or inhibitory signals are established. Immune checkpoint inhibitors (ICIs), block these interactions, and when administered not only as monotherapy but also as part of combination regimens, have shown to improve survival results in multiple advanced cancers leading to an increasing number of patients treated with ICI and, as a consequence, a rise in the number of patients developing immune-related adverse events (irAEs). Presence of irAEs has been associated with greater benefit from treatment, especially when blocking PD-L1. Recent data suggests that treatment benefit persists after discontinuation of ICIs due to a treatment related adverse event, regardless of the grade. Patients experiencing grade 3-4 irAEs are at risk of toxicity recurrence after reintroducing immunotherapy and therefore, the decision to resume the treatment is challenging. In these cases, a multidisciplinary approach is always needed and several factors should be considered. Management of severe toxicities may require systemic corticosteroids which can impact on T-cell function. Due to their immunosuppressive properties, it is necessary to deeper determine how corticosteroids influence responses. In terms of overall survival (OS), the use of steroids as therapy for irAEs seems not to reduce OS and several studies have reported durable responses in patients experiencing autoimmune toxicities treated with corticosteroids.
Collapse
Affiliation(s)
- Patricia Iranzo
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Ana Callejo
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Juan David Assaf
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Gaspar Molina
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Daniel Esteban Lopez
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - David Garcia-Illescas
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Nuria Pardo
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Alejandro Navarro
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Alex Martinez-Marti
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Susana Cedres
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Caterina Carbonell
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Joan Frigola
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ramon Amat
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
89
|
Navani V, Graves MC, Mandaliya H, Hong M, van der Westhuizen A, Martin J, Bowden NA. Melanoma: An immunotherapy journey from bench to bedside. Cancer Treat Res 2022; 183:49-89. [PMID: 35551656 DOI: 10.1007/978-3-030-96376-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Melanoma gave science a window into the role immune evasion plays in the development of malignancy. The entire spectrum of immune focused anti-cancer therapies has been subjected to clinical trials in this disease, with limited success until the immune checkpoint blockade era. That revolution launched first in melanoma, heralded a landscape change throughout cancer that continues to reverberate today.
Collapse
Affiliation(s)
| | - Moira C Graves
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia
| | - Hiren Mandaliya
- Calvary Mater Hospital Newcastle, Edith St, Waratah, NSW, 2298, Australia
| | - Martin Hong
- Calvary Mater Hospital Newcastle, Edith St, Waratah, NSW, 2298, Australia
| | - Andre van der Westhuizen
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia.,Calvary Mater Hospital Newcastle, Edith St, Waratah, NSW, 2298, Australia
| | - Jennifer Martin
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia.,John Hunter Hospital, Newcastle, NSW, Australia
| | - Nikola A Bowden
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia
| |
Collapse
|
90
|
Alvarez-Breckenridge C, Remon J, Piña Y, Nieblas-Bedolla E, Forsyth P, Hendriks L, Brastianos PK. Emerging Systemic Treatment Perspectives on Brain Metastases: Moving Toward a Better Outlook for Patients. Am Soc Clin Oncol Educ Book 2022; 42:1-19. [PMID: 35522917 DOI: 10.1200/edbk_352320] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The diagnosis of brain metastases has historically been a dreaded, end-stage complication of systemic disease. Additionally, with the increasing effectiveness of systemic therapies that prolong life expectancy and improved imaging tools, the incidence of intracranial progression is becoming more common. Within this context, there has been increasing attention directed at understanding the molecular underpinnings of intracranial progression. Exploring the unique features of brain metastases compared with their extracranial counterparts to identify aberrant signaling pathways, which can be targeted pharmacologically, may help lead to new treatments for this patient population. Additionally, critical discoveries outside the sphere of the central nervous system are increasingly being applied to brain metastases with the emergence of immune checkpoint inhibition, becoming a prevalent treatment option for patients with brain metastases across multiple histologies. As novel treatment strategies are considered, they require thoughtful incorporation of agents that can cross the blood-brain barrier and can synergize with pre-existing agents through rational combinations. Lastly, as clinicians and scientists continue to understand key molecular features of these tumors, they will continue to influence the treatment algorithms that are developing for the management of these patients. Due to the complexity of treatment decisions for patients with brain metastases, an emerging tool is the utilization of multidisciplinary brain metastasis tumor boards to ensure optimal treatment decisions are made and that patients are provided access to applicable clinical trials. Looking to the future, the collective effort to understand the various tumor-intrinsic and tumor-extrinsic factors that promote central nervous system seeding and propagation will have the potential to change the clinical trajectory for these patients.
Collapse
Affiliation(s)
| | - Jordi Remon
- Department of Medical Oncology, HM CIOCC Barcelona (Centro Integral Oncológico Clara Campal), Hospital HM Delfos, HM Hospitales, Barcelona, Spain
| | - Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL
| | | | - Peter Forsyth
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL
| | - Lizza Hendriks
- Department of Pulmonary Diseases - GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, Netherlands
| | | |
Collapse
|
91
|
Xavier CB, Lopes CDH, Harada G, Peres EDB, Katz A, Jardim DL. Cardiovascular toxicity following immune checkpoint inhibitors: A systematic review and meta-analysis. Transl Oncol 2022; 19:101383. [PMID: 35248919 PMCID: PMC8898968 DOI: 10.1016/j.tranon.2022.101383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/10/2022] [Accepted: 02/23/2022] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors may be associated with multiple immune-related toxicities. Cardiovascular adverse effects are underreported in clinical trials. METHODS We conducted a systematic review and meta-analysis to evaluate cardiovascular adverse effects incidence among patients with solid tumors receiving immune checkpoint inhibitors in randomized clinical trials and the relative risk of presenting these effects compared to placebo or best supportive care. The search was conducted through MEDLINE, Embase, and Scopus databases from January 1st, 2010 until July 1st, 2020. Outcomes were reported following Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. RESULTS 57 randomized clinical trials including 12,118 patients were included. All grade CV AEs incidence rate was 8.32% (95% CI = 6.35%-10.53%). When only grade 3-5 CV AEs were considered, ICIs were significantly associated with increased risk than placebo or BSC (RR = 1.36; 95% CI = 1.06-1.73; p = 0.01). CONCLUSION This meta-analysis corroborates the hypothesis of increased CV risk related to immune checkpoint inhibitors.
Collapse
Key Words
- Adverse effects, BSC
- Best supportive care, CTCAE
- Cardiovascular toxicity
- Cardiovascular, FAERS
- Checkpoint blockade
- Common toxicity criteria, CTLA-4
- Cytotoxic t-lymphocyte-associated antigen 4, CV
- Immune checkpoint inhibitors
- Immune checkpoint inhibitors, PRISMA
- Meta-analysis, abbreviations, AES
- Preferred Reporting Items for Systematic Reviews and Meta-analyses, RCTs
- U.S. food and drug administration adverse events reporting system, ICIS
- programmed cell death ligand 1, RR
- programmed cell death protein 1, PD-L1
- randomized clinical trials, PD-1
- relative risk
Collapse
Affiliation(s)
| | | | | | | | - Artur Katz
- Oncology Center - Hospital Sírio-Libanês, São Paulo, Brazil
| | | |
Collapse
|
92
|
Abstract
Immune checkpoint inhibitors (ICIs) have been recently proposed as a strategy for treating anti-malignant neoplasms. However, this treatment leads to immune-related adverse events (irAEs) such as autoimmune endocrinopathy. Early diagnosis and appropriate treatment of ICI-related hypophysitis are essential as it can manifest as a life-threatening condition due to an adrenal crisis. In this review, we summarize the pathogenesis, risk factors, diagnostic processes, clinical characteristics, and its current management. In particular, we discuss the different aspects of anti-CTLA-4 antibody-related and anti-PD-1/anti-PD-L1 antibody-related hypophysitis. We also propose key points for early detection and diagnosis by identifying the target group that should be monitored more carefully. Specific methods of hormone replacement therapy have also been described. We hope that this review will lead to a better understanding and management of this rare but serious condition during cancer treatment and further elucidate the pathophysiology of pituitary autoimmunity.
Collapse
Affiliation(s)
- Tadashi Mizukoshi
- Kobe University School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Kobe University Hospital, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Yutaka Takahashi
- Kobe University School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; Department of Diabetes and Endocrinology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| |
Collapse
|
93
|
Switzer B, Puzanov I, Skitzki JJ, Hamad L, Ernstoff MS. Managing Metastatic Melanoma in 2022: A Clinical Review. JCO Oncol Pract 2022; 18:335-351. [PMID: 35133862 PMCID: PMC9810138 DOI: 10.1200/op.21.00686] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cutaneous melanoma remains the most lethal of the primary cutaneous neoplasms, and although the incidence of primary melanoma continues to rise, the mortality from metastatic disease remains unchanged, in part through advances in treatment. Major developments in immunomodulatory and targeted therapies have provided robust improvements in response and survival trends that have transformed the clinical management of patients with metastatic melanoma. Additional advances in immunologic and cancer cell biology have contributed to further optimization in (1) risk stratification, (2) prognostication, (3) treatment, (4) toxicity management, and (5) surveillance approaches for patients with an advanced melanoma diagnosis. In this review, we provide a comprehensive overview of the historical and future advances regarding the translational and clinical implications of advanced melanoma and share multidisciplinary recommendations to aid clinicians in the navigation of current treatment approaches for a variety of patient cohorts.
Collapse
Affiliation(s)
- Benjamin Switzer
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Joseph J. Skitzki
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Lamya Hamad
- Department of Pharmacy, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Marc S. Ernstoff
- ImmunoOncology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD,Marc S. Ernstoff, MD, National Cancer Institute, Rockville, MD 20850; e-mail:
| |
Collapse
|
94
|
Araujo DV, Uchoa B, Soto-Castillo JJ, Furlan LL, Oliva M. When Less May Be Enough: Dose Selection Strategies for Immune Checkpoint Inhibitors Focusing on AntiPD-(L)1 Agents. Target Oncol 2022; 17:253-270. [PMID: 35687223 DOI: 10.1007/s11523-022-00890-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
Abstract
Early clinical trials investigating antiPD(L)-1 agents rarely reached a maximum tolerated dose (MTD), and efficacy signals were observed even at the lowest dose levels. Most extended treatment intervals investigated indicated that these drugs do not follow a direct dose-toxicity or dose-efficacy relationship. Within this context and considering the high cost of antiPD(L)-1 agents, there is a significant debate on whether lower doses or the administration of such agents at an extended interval should be prospectively evaluated in already-approved agents, or at least be considered in novel combination trials involving antiPD(L)-1 drugs. Herein, we review the dosing, overall response rates, and incidence of treatment-related adverse events of antiPD(L)-1 agents in early dose-escalation trials and discuss the appropriateness of recommended Phase 2 dose selection as well as the final regulatory approved doses of such agents. Efficacy and safety data from randomized dose-range Phase 2 trials and real-world data (RWD) on the usage of lower doses and/or non-standard extended treatment intervals are also examined. As the accumulating evidence suggests lower doses or extended dosing intervals of antiPD(L)-1 may achieve a similar clinical benefit in comparison to the currently approved doses, we address the clinical and financial toxicity implications of using potentially higher doses than necessary. Last, we discuss ways to resolve the current dosing conundrum of antiPD-(L)1 agents such as performing near-equivalence studies and propose a framework for future development of immunotherapeutics to find the lowest efficacious dose instead of MTD.
Collapse
Affiliation(s)
- Daniel V Araujo
- Department of Medical Oncology, Hospital de Base/HB Onco, FUNFARME/FAMERP, Av. Brigadeiro Faria Lima 5544, São José do Rio Preto, SP, Brazil.
| | - Bruno Uchoa
- Department of Medical Oncology, Hospital de Base/HB Onco, FUNFARME/FAMERP, Av. Brigadeiro Faria Lima 5544, São José do Rio Preto, SP, Brazil
| | - Juan José Soto-Castillo
- Department of Medical Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Av. Gran Via de L'Hospitalet 199-203, 08908, Barcelona, Spain
| | - Larissa L Furlan
- Department of Medical Oncology, Hospital de Base/HB Onco, FUNFARME/FAMERP, Av. Brigadeiro Faria Lima 5544, São José do Rio Preto, SP, Brazil
| | - Marc Oliva
- Department of Medical Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Av. Gran Via de L'Hospitalet 199-203, 08908, Barcelona, Spain. .,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
95
|
Yuen KCJ, Samson SL, Bancos I, Gosmanov AR, Jasim S, Fecher LA, Weber JS. AMERICAN ASSOCIATION OF CLINICAL ENDOCRINOLOGY (AACE) DISEASE STATE CLINICAL REVIEW EVALUATION AND MANAGEMENT OF IMMUNE CHECKPOINT INHIBITOR-MEDIATED ENDOCRINOPATHIES: A PRACTICAL CASE-BASED CLINICAL APPROACH. Endocr Pract 2022; 28:719-731. [PMID: 35477029 DOI: 10.1016/j.eprac.2022.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The aim of this case-based clinical review is to provide a practical approach for clinicians regarding the management of patients with immune checkpoint inhibitor (ICI)-mediated endocrinopathies. METHODS A literature search was conducted using PubMed, Embase and Scopus, and appropriate keywords. The discussions and strategies for diagnosis and management of ICI-mediated endocrinopathies are based on evidence available from prospective randomized clinical studies, cohort studies, cross-sectional studies, case-based studies, and expert consensus. RESULTS Immunotherapy with ICIs has transformed the treatment landscape of diverse cancer types, but frequently results in immune-mediated endocrinopathies that can cause acute and persistent morbidity, and rarely, death. The patterns of endocrinopathies differ between inhibitors of the CTLA-4 and PD-1/PD-L1 pathways, but most often involve the thyroid and pituitary glands. Less common but important presentations include insulin-deficient diabetes mellitus, primary adrenal insufficiency, primary hypoparathyroidism, central diabetes insipidus, primary hypogonadism, and pancreatitis with or without subsequent progression to diabetes or exocrine insufficiency. CONCLUSION In recent years, with increasing numbers of cancer patients being treated with ICIs, more clinicians in a variety of specialties are called upon to diagnose and treat ICI-mediated endocrinopathies. Herein, we review case scenarios of various clinical manifestations, and emphasize the need for a high index of clinical suspicion by all clinicians caring for these patients including endocrinologists, oncologists, primary care providers, and emergency department physicians. We also provide diagnostic and therapeutic approaches for ICI-induced endocrinopathies, and we propose that patients on ICI-therapy be evaluated and treated in a multidisciplinary team in collaboration with endocrinologists.
Collapse
Affiliation(s)
- Kevin C J Yuen
- Co-Chair of Task Force; Professor of Medicine, Department of Medicine, University of Arizona College of Medicine and Creighton School of Medicine, Phoenix, Arizona.
| | - Susan L Samson
- Co-Chair of Task Force; Senior Associate Consultant, Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Jacksonville, Florida
| | - Irina Bancos
- Associate Professor of Medicine; Associate Program Director, Endocrinology Fellowship Program, Division of Endocrinology, Metabolism and Nutrition, Mayo Clinic, Rochester, MN
| | - Aidar R Gosmanov
- Professor of Medicine, Division of Endocrinology, Albany Medical College; Chief, Endocrinology Section, Stratton VAMC, Albany, NY
| | - Sina Jasim
- Associate Professor of Medicine, Washington University in St. Louis, School of Medicine, Division of Endocrinology, Metabolism and Lipid Research, St. Louis, Missouri
| | - Leslie A Fecher
- ASCO Representative, Associate Professor of Medicine and Dermatology, University of Michigan, Rogel Cancer Center, Ann Arbor, Michigan
| | - Jeffrey S Weber
- ASCO Representative, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|
96
|
Goel B, Tiwari AK, Pandey RK, Singh AP, Kumar S, Sinha A, Jain SK, Khattri A. Therapeutic approaches for the treatment of head and neck squamous cell carcinoma-An update on clinical trials. Transl Oncol 2022; 21:101426. [PMID: 35460943 PMCID: PMC9046875 DOI: 10.1016/j.tranon.2022.101426] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common non-skin cancer with a tobacco consumption and infection with high-risk human papillomavirus (HPV) being major risk factors. Despite advances in numerous therapy modalities, survival rates for HNSCC have not improved considerably; a vast number of clinical outcomes have demonstrated that a combination strategy (the most well-known docetaxel, cisplatin, and 5-fluorouracil) is the most effective treatment choice. Immunotherapy that targets immunological checkpoints is being tested in a number of clinical trials, either alone or in conjunction with chemotherapeutic or targeted therapeutic drugs. Various monoclonal antibodies, such as cetuximab and bevacizumab, which target the EGFR and VEGFR, respectively, as well as other signaling pathway inhibitors, such as temsirolimus and rapamycin, are also being studied for the treatment of HNSCC. We have reviewed the primary targets in active clinical studies in this study, with a particular focus on the medications and drug targets used.
Collapse
Affiliation(s)
- Bharat Goel
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Anoop Kumar Tiwari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Rajeev Kumar Pandey
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Akhand Pratap Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Sujeet Kumar
- Centre for Proteomics and Drug Discovery, Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai - 410206, Maharashtra, India
| | - Abhishek Sinha
- Department of Oral Medicine & Radiology, Sardar Patel Post Graduate Institute of Dental & Medical Sciences, Lucknow - 226025, Uttar Pradesh, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India
| | - Arun Khattri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India.
| |
Collapse
|
97
|
Evangelista GFDB, Figueiredo AB, Barros e Silva MJD, Gollob KJ. Balancing the good and the bad: controlling immune related adverse events versus anti-tumor responses in cancer patients treated with immune checkpoint inhibitors. IMMUNOTHERAPY ADVANCES 2022; 2:ltac008. [PMID: 35919497 PMCID: PMC9327097 DOI: 10.1093/immadv/ltac008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) have provided new hope for cancer patients, and in particular for patients with tumors that are immunologically active and classified as hot tumors. These tumors express antigenic and tumor microenvironment (TME) characteristics that make them potential candidates for therapy with checkpoint inhibitors that aim to reactivate the immune response such as anti-PD-1 and anti-CTLA-4. Examples of potentially responsive cancers are, melanoma, non-small cell lung cancer and several other metastatic or unresectable tumors with genetic instability: DNA mismatch repair deficiency (dMMR), microsatellite instability-high (MSI-H), or with a high tumor mutational burden (TMB). Immunotherapy using checkpoint inhibitors is typically associated with adverse events (AEs) that are milder than those with chemotherapy. However, a significant percentage of patients develop short-term immune-related AEs (irAEs) which range from mild (~70%) to severe cases (~13%) that can lead to modifications of the checkpoint inhibitor therapy and in some cases, death. While some studies have investigated immune mechanisms behind the development of irAEs, much more research is needed to understand the mechanisms and to develop interventions that could attenuate severe irAEs, while maintaining the anti-tumor response intact. Moreover, studies to identify biomarkers that can predict the likelihood of a patient developing severe irAEs would be of great clinical importance. Here we discuss some of the clinical ramifications of irAEs, potential immune mechanisms behind their development and studies that have investigated potentially useful biomarkers of irAEs development.
Collapse
Affiliation(s)
- Guilherme Ferreira de Britto Evangelista
- Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Center for Research in Immuno-oncology (CRIO), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Translational Immuno-oncology Group, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Amanda Braga Figueiredo
- Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Center for Research in Immuno-oncology (CRIO), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Kenneth J Gollob
- Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Center for Research in Immuno-oncology (CRIO), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| |
Collapse
|
98
|
Hong WX, Sagiv-Barfi I, Czerwinski DK, Sallets A, Levy R. Neoadjuvant Intratumoral Immunotherapy with TLR9 Activation and Anti-OX40 Antibody Eradicates Metastatic Cancer. Cancer Res 2022; 82:1396-1408. [PMID: 35135810 PMCID: PMC8983569 DOI: 10.1158/0008-5472.can-21-1382] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/01/2021] [Accepted: 02/04/2022] [Indexed: 11/16/2022]
Abstract
The combination of the synthetic TLR9 ligand CpG and agnostic OX40 antibody can trigger systemic antitumor immune responses upon co-injection into the tumor microenvironment, eradicating simultaneous untreated sites of metastatic disease. Here we explore the application of this in situ immunotherapy to the neoadjuvant setting. Current neoadjuvant checkpoint blockade therapy is delivered systemically, resulting in off-target adverse effects. In contrast, intratumoral immunotherapy minimizes the potential for toxicities and allows for greater development of combination therapies. In two metastatic solid tumor models, neoadjuvant intratumoral immunotherapy generated a local T-cell antitumor response that then acted systemically to attack cancer throughout the body. In addition, the importance of timing between neoadjuvant immunotherapy and surgical resection was established, as well as the increased therapeutic power of adding systemic anti-PD1 antibody. The combination of local and systemic immunotherapy generated an additional survival benefit due to synergistic inhibitory effect on tumor-associated macrophages. These results provide a strong rationale for translating this neoadjuvant intratumoral immunotherapy to the clinical setting, especially in conjunction with established checkpoint inhibitors. SIGNIFICANCE This work demonstrates the ability of neoadjuvant intratumoral immunotherapy to target local and distant metastatic disease and consequently improve survival.
Collapse
Affiliation(s)
- Wan Xing Hong
- Department of Surgery, Stanford University School of Medicine, Department of Medicine, Stanford University
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University
| | - Idit Sagiv-Barfi
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University
| | - Debra K. Czerwinski
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University
| | - Adrienne Sallets
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University
| | - Ronald Levy
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University
| |
Collapse
|
99
|
Song G, Zhang FF, Cheng HD. Thalidomide for prevention of camrelizumab-induced reactive cutaneous capillary endothelial proliferation. Australas J Dermatol 2022; 63:217-221. [PMID: 35229882 PMCID: PMC9313837 DOI: 10.1111/ajd.13812] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 01/10/2023]
Abstract
Objectives The study evaluated the efficacy of thalidomide in prevention of camrelizumab‐induced reactive cutaneous capillary endothelial proliferation (RCCEP). Methods In this study, patients treated with camrelizumab plus thalidomide or camrelizumab alone were included. The occurrences, onset time, severity of RCCEP and the adverse effect of thalidomide were analysed. Results A total of 19 patients were enrolled. The incidence of RCCEP in thalidomide group (2/9, 22.2%) was significantly lower than that in camrelizumab group (8/10, 80%). The median onset time of RCCEP was 5 weeks and 4 weeks respectively. The adverse events of thalidomide were mild, and no treatment‐associated interruption was observed. Conclusions Thalidomide showed a promising in prevention of the RCCEP in patients receiving camrelizumab therapy with an acceptable safety profile.
Collapse
Affiliation(s)
- Geng Song
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fei-Fei Zhang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huai-Dong Cheng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
100
|
Schonfeld SJ, Tucker MA, Engels EA, Dores GM, Sampson JN, Shiels MS, Chanock SJ, Morton LM. Immune-Related Adverse Events After Immune Checkpoint Inhibitors for Melanoma Among Older Adults. JAMA Netw Open 2022; 5:e223461. [PMID: 35315916 PMCID: PMC8941351 DOI: 10.1001/jamanetworkopen.2022.3461] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/31/2022] [Indexed: 12/17/2022] Open
Abstract
Importance Immune checkpoint inhibitors (ICIs) have improved survival in patients with advanced melanoma but can be associated with a spectrum of immune-related adverse events (AEs), including both autoimmune-related AEs and other immune-related inflammatory AEs. These associations have primarily been evaluated in clinical trials that include highly selected patients, with older adults often underrepresented. Objective To evaluate the association between use of ICIs and immune-related AEs (autoimmune and other immune related) among older patients with cutaneous melanoma. Design, Setting, and Participants A population-based cohort study was conducted from January 1, 2011, to December 31, 2015. Data were analyzed from January 31 to May 31, 2021. With use of a linked database of Medicare claims and Surveillance, Epidemiology, and End Results (SEER) Program population-based cancer registries, patients of White race diagnosed with stages II-IV or unknown (American Joint Committee on Cancer, AJCC Cancer Staging Manual 6th edition) first primary cutaneous melanoma during 2011-2015, as reported to SEER, and followed up through December 31, 2015, were identified. Exposures Immune checkpoint inhibitors for treatment of melanoma. Main Outcomes and Measures The association between ICIs and immune-related AEs ascertained from Medicare claims data was estimated using multivariable Cox regression with hazard ratios (HRs) and 95% CIs and with cumulative incidence accounting for competing risk of death. Results The study included 4489 patients of White race with first primary melanoma (3002 men [66.9%]; median age, 74.9 [range, 66.0-84.9] years). During follow-up (median, 1.4 [range, 0-5.0] years), 1576 patients (35.1%) had an immune-related AE on a Medicare claim. Use of ICIs (reported for 418 patients) was associated with autoimmune-related AEs (HR, 2.5; 95% CI, 1.6-4.0), including primary adrenal insufficiency (HR, 9.9; 95% CI, 4.5-21.5) and ulcerative colitis (HR, 8.6; 95% CI, 2.8-26.3). Immune checkpoint inhibitors also were associated with other immune-related AEs (HR, 2.2; 95% CI, 1.7-2.8), including Cushing syndrome (HR, 11.8; 95% CI, 1.4-97.2), hyperthyroidism (HR, 6.3; 95% CI, 2.0-19.5), hypothyroidism (HR, 3.8; 95% CI, 2.4-6.1), hypopituitarism (HR, 19.8; 95% CI, 5.4-72.9), other pituitary gland disorders (HR, 6.0; 95% CI, 1.2-30.2), diarrhea (HR, 3.5; 95% CI, 2.5-4.9), and sepsis or septicemia (HR, 2.2; 95% CI, 1.4-3.3). Most associations were pronounced within 6 months following the first ICI claim and comparable with or without a baseline history of autoimmune disease. The cumulative incidence at 6 months following the first ICI claim was 13.7% (95% CI, 9.7%-18.3%) for autoimmune-related AEs and 46.8% (95% CI, 40.7%-52.7%) for other immune-related AEs. Conclusions and Relevance In this cohort study of older adults with melanoma, ICIs were associated with autoimmune-related AEs and other immune-related AEs. Although some findings were consistent with clinical trials of ICIs, others warrant further investigation. As ICI use continues to expand rapidly, ongoing investigation of the spectrum of immune-related AEs may optimize management of disease in patients.
Collapse
Affiliation(s)
- Sara J. Schonfeld
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Margaret A. Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Eric A. Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Graça M. Dores
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Joshua N. Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Meredith S. Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Lindsay M. Morton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| |
Collapse
|