1
|
Maul LV, Ramelyte E, Dummer R, Mangana J. Management of metastatic melanoma with combinations including PD-1 inhibitors. Expert Opin Biol Ther 2025:1-12. [PMID: 40159098 DOI: 10.1080/14712598.2025.2485315] [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: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Melanoma is among the most immunogenic malignancies. The advent of immune checkpoint inhibitors (ICIs) has revolutionized the landscape of melanoma treatment. Long-term durable cancer control is possible in nearly 50% of non-resectable, metastatic melanoma patients with anti-CTLA4 and anti-PD-1 antibodies. AREAS COVERED This review provides a critical overview of the current data and future research directions on the management of metastatic melanoma with ICIs. We reviewed the efficacy and safety of combinations with PD-1 inhibitors through PubMed database research (Nov 2024-Mar 2025). EXPERT OPINION A decade after ipilimumab's approval, challenges remain. To cure more patients, the development of combinations is warranted. Combinations with a limited number of ipilimumab applications improve the overall survival outcome by approximately 10%, with a dramatic increase in adverse events including fatal events. Anti-LAG3/nivolumab is a promising alternative, offering similar efficacy to ipilimumab/nivolumab with better tolerability. In our opinion, ipilimumab/nivolumab combination should be the first-line therapy for high-risk patients (high LDH, brain or liver metastasis), while nivolumab/relatlimab or PD-1 monotherapy may be preferable for lower-risk cases. However, treatment decisions are increasingly complex, since most patients nowadays are pretreated in the (neo)-adjuvant setting. The key limitation today is the lack of biomarkers to guide individualized treatment strategies.
Collapse
Affiliation(s)
- Lara Valeska Maul
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Huang RY, Youssef G, Nelson T, Wen PY, Forsyth P, Hodi FS, Margolin K, Algazi AP, Hamid O, Lao CD, Ernstoff MS, Moschos SJ, Atkins MB, Postow MA, Reardon DA, Grootendorst DJ, Leung D, Askelson M, Ritchings C, Tawbi HA. Comparative Analysis of Intracranial Response Assessment Criteria in Patients With Melanoma Brain Metastases Treated With Combination Nivolumab + Ipilimumab in CheckMate 204. J Clin Oncol 2025; 43:1210-1218. [PMID: 39752606 PMCID: PMC11949218 DOI: 10.1200/jco.24.00953] [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: 05/02/2024] [Revised: 10/24/2024] [Accepted: 11/18/2024] [Indexed: 03/29/2025] Open
Abstract
PURPOSE In CheckMate 204, nivolumab + ipilimumab showed high intracranial (IC) objective response rates (icORRs) in patients with melanoma brain metastases (MBMs). Using icORR as a surrogate for overall survival (OS) has prompted use of alternate response criteria. To set the stage for harmonized MBM trials, the aim of this exploratory analysis was to determine icORR using several response criteria and examine correlations of response with survival. METHODS Patients (N = 119) with ≥one unirradiated MBMs received nivolumab + ipilimumab every 3 weeks (four doses), followed by nivolumab every 2 weeks for ≤24 months. Blinded review icORR was assessed with modified RECIST (mRECIST), Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM; 5 mm and 10 mm cutoffs), and volumetric criteria (5 mm and 10 mm). Using a 6-week response landmark, IC progression-free survival (icPFS) and OS were compared for responders versus nonresponders. RESULTS icORR was higher with mRECIST and volumetric criteria than with RANO-BM or RECIST. mRECIST and volumetric response also showed stronger correlations with icPFS and OS. mRECIST responders who were RANO-BM 5 mm nonresponders (n = 14) had similar OS to RANO-BM 5 mm responders (n = 41). Clinical deterioration affected RANO-BM icORR; however, when assessed only radiographically without deterioration, RANO-BM 5 mm performed similarly to mRECIST. Among 41 patients with target lesions all <10 mm, responder icPFS and OS were similar to those of responders in the total population, indicating that response could be accurately determined in these patients. CONCLUSION This analysis supports mRECIST or radiographic-only RANO-BM 5 mm as reliable assessment scales in MBM trials. Volumetric response correlated with survival, supporting its application in future trials. Response could be accurately determined in patients with MBMs all <10 mm, supporting the inclusion of patients with MBMs ≥5 mm in future trials.
Collapse
Affiliation(s)
- Raymond Y. Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gilbert Youssef
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Thomas Nelson
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Patrick Y. Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Peter Forsyth
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL
| | - F. Stephen Hodi
- Medical Oncology, Dana-Farber Cancer Institute, Massachusetts General Hospital, Boston, MA
| | - Kim Margolin
- Department of Medical Oncology, Saint John's Cancer Institute, Santa Monica, CA
| | - Alain P. Algazi
- Department of Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - Omid Hamid
- The Angeles Clinic and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, CA
| | - Christopher D. Lao
- Department of Dermatology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Marc S. Ernstoff
- Department of Immuno-Oncology, Division of Cancer Treatment and Diagnosis, National Cancer Institute at the National Institutes of Health, Rockville, MD
| | - Stergios J. Moschos
- Division of Medical Oncology, The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Michael B. Atkins
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC
| | - Michael A. Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - David A. Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | | | | | - Hussein A. Tawbi
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
3
|
Rajczykowski M, Olbryt M, Galwas K, Idasiak A, Stobiecka E, Suwiński R. Sustained Immunotherapy Response in Metastatic Brain Melanoma Through 2 Pregnancies. AMERICAN JOURNAL OF CASE REPORTS 2025; 26:e945533. [PMID: 40083082 PMCID: PMC11918453 DOI: 10.12659/ajcr.945533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
BACKGROUND Metastatic brain melanoma is a deadly form of cancer with a high mortality rate and short overall survival. Immunotherapy with immune checkpoint inhibitors is the first treatment option for BRAF wild-type patients. Pregnancy is the exclusion criterion for immunotherapy and may promote the progression of melanoma. This report shows the long-lasting response of a patient with metastasis in multiple locations, including the brain, to immunotherapy and radiotherapy, who delivered 2 healthy boys during the disease. CASE REPORT A 39-year-old woman was diagnosed with BRAF(-)/NRAS(+) skin melanoma, pT2bN2aM0 (IIIB). Due to pregnancy, she did not receive adjuvant therapy. Upon delivery, the disease manifested with multiple extracranial and symptomatic brain metastasis. She was treated with whole-brain radiation and immunotherapy with ipilimumab and nivolumab followed by nivolumab. A partial response of the brain metastases and an extracranial complete response were observed. During the immunotherapy, she became pregnant and the therapy was discontinued. She was under regular medical surveillance, during which she delivered a healthy boy. The last CT scan and magnetic resonance brain examination showed a maintenance response for 43 months after initiation of immunotherapy and 31 months after therapy completion. CONCLUSIONS A long-lasting response to radiotherapy and interrupted immunotherapy is possible in the case of symptomatic metastatic brain melanoma developing during pregnancy, and healthy deliveries are possible despite the mother's progressive melanoma or exposure of the fetus to nivolumab (first trimester).
Collapse
Affiliation(s)
- Marcin Rajczykowski
- II Radiotherapy and Chemotherapy Clinic and Teaching Hospital, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Magdalena Olbryt
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Katarzyna Galwas
- II Radiotherapy and Chemotherapy Clinic and Teaching Hospital, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Adam Idasiak
- II Radiotherapy and Chemotherapy Clinic and Teaching Hospital, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Ewa Stobiecka
- Department of Tumor Pathology, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Rafał Suwiński
- II Radiotherapy and Chemotherapy Clinic and Teaching Hospital, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| |
Collapse
|
4
|
Rodriguez-Baena FJ, Marquez-Galera A, Ballesteros-Martinez P, Castillo A, Diaz E, Moreno-Bueno G, Lopez-Atalaya JP, Sanchez-Laorden B. Microglial reprogramming enhances antitumor immunity and immunotherapy response in melanoma brain metastases. Cancer Cell 2025; 43:413-427.e9. [PMID: 39919736 DOI: 10.1016/j.ccell.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/04/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Melanoma is one of the tumor types with the highest risk of brain metastasis. However, the biology of melanoma brain metastasis and the role of the brain immune microenvironment in treatment responses are not yet fully understood. Using preclinical models and single-cell transcriptomics, we have identified a mechanism that enhances antitumor immunity in melanoma brain metastasis. We show that activation of the Rela/Nuclear Factor κB (NF-κB) pathway in microglia promotes melanoma brain metastasis. Targeting this pathway elicits microglia reprogramming toward a proinflammatory phenotype, which enhances antitumor immunity and reduces brain metastatic burden. Furthermore, we found that proinflammatory microglial markers in melanoma brain metastasis are associated with improved responses to immune checkpoint inhibitors in patients and targeting Rela/NF-κB pathway in mice improves responses to these therapies in the brain, suggesting a strategy to enhance antitumor immunity and responses to immune checkpoint inhibitors in patients with melanoma brain metastasis.
Collapse
Affiliation(s)
| | | | | | - Alba Castillo
- Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, Spain
| | - Eva Diaz
- MD Anderson Cancer Center International Foundation, Madrid, Spain
| | - Gema Moreno-Bueno
- MD Anderson Cancer Center International Foundation, Madrid, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" CSIC-UAM, Madrid, Spain; CIBERONC Centro de Investigación Biomédica en Red de Cancer, ISCIII, Madrid, Spain; Translational Cancer Research Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | | | | |
Collapse
|
5
|
Ding J, Jiang Y, Jiang N, Xing S, Ge F, Ma P, Tang Q, Miao H, Zhou J, Fang Y, Cui D, Liu D, Han Y, Yu W, Wang Y, Zhao G, Cai Y, Wang S, Sun N, Li N. Bridging the gap: unlocking the potential of emerging drug therapies for brain metastasis. Brain 2025; 148:702-722. [PMID: 39512184 DOI: 10.1093/brain/awae366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/30/2024] [Accepted: 09/29/2024] [Indexed: 11/15/2024] Open
Abstract
Brain metastasis remains an unmet clinical need in advanced cancers with an increasing incidence and poor prognosis. The limited response to various treatments is mainly derived from the presence of the substantive barrier, blood-brain barrier (BBB) and brain-tumour barrier (BTB), which hinders the access of potentially effective therapeutics to the metastatic tumour of the brain. Recently, the understanding of the structural and molecular features of the BBB/BTB has led to the development of efficient strategies to enhance BBB/BTB permeability and deliver drugs across the BBB/BTB to elicit the anti-tumour response against brain metastasis. Meanwhile, novel agents capable of penetrating the BBB have rapidly developed and been evaluated in preclinical studies and clinical trials, with both targeted therapies and immunotherapies demonstrating impressive intracranial activity against brain metastasis. In this review, we summarize the recent advances in the biological properties of the BBB/BTB and the emerging strategies for BBB/BTB permeabilization and drug delivery across the BBB/BTB. We also discuss the emerging targeted therapies and immunotherapies against brain metastasis tested in clinical trials. Additionally, we provide our viewpoints on accelerating clinical translation of novel drugs into clinic for patients of brain metastasis. Although still challenging, we expect this review to benefit the future development of novel therapeutics, specifically from a clinical perspective.
Collapse
Affiliation(s)
- Jiatong Ding
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yale Jiang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Jiang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shujun Xing
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Fan Ge
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Peiwen Ma
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiyu Tang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Huilei Miao
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiawei Zhou
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuan Fang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dandan Cui
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dongyan Liu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yanjie Han
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Weijie Yu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuning Wang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guo Zhao
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuanting Cai
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuhang Wang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
6
|
Long GV, Atkinson V, Lo SN, Guminski AD, Sandhu SK, Brown MP, Gonzalez M, McArthur GA, Menzies AM. Ipilimumab plus nivolumab versus nivolumab alone in patients with melanoma brain metastases (ABC): 7-year follow-up of a multicentre, open-label, randomised, phase 2 study. Lancet Oncol 2025; 26:320-330. [PMID: 39978375 DOI: 10.1016/s1470-2045(24)00735-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND Patients with melanoma brain metastases respond well to immunotherapy, but long-term comparative survival data are scarce. We aimed to assess the efficacy of ipilimumab plus nivolumab versus nivolumab alone in patients with melanoma brain metastases at 7 years. METHODS This open-label, randomised, phase 2 study was conducted at four sites (two research institute cancer centres and two university teaching hospitals) in Australia. Patients aged 18 years or older with active, immunotherapy-naive melanoma brain metastases and Eastern Cooperative Oncology Group performance status of 0-2 were eligible. Asymptomatic patients with no previous brain-directed therapy were randomly assigned (5:4) using the biased-coin minimisation method (after a safety run-in of six patients) to cohort A (intravenous ipilimumab 3 mg/kg plus nivolumab 1 mg/kg every 3 weeks for four doses, then nivolumab 3 mg/kg every 2 weeks) or cohort B (intravenous nivolumab 3 mg/kg every 2 weeks). Patients with previous brain-directed therapy, neurological symptoms, or leptomeningeal disease were assigned to cohort C (non-randomised; intravenous nivolumab 3 mg/kg every 2 weeks). The primary endpoint was best intracranial response (complete or partial response) from week 12. Secondary survival endpoints included intracranial progression-free survival and overall survival. Safety was assessed from the first dose of treatment to at least 100 days after treatment discontinuation. Analyses were performed in patients who received at least one dose of study drug. The main analysis has been reported, and this is a long-term follow up of the ABC trial. This trial is registered with ClinicalTrials.gov, NCT02374242, and is ongoing. FINDINGS Between Nov 4, 2014, and April 21, 2017, 89 patients were assessed for eligibility, 79 of whom were enrolled and assigned to cohort A (n=36), cohort B (n=27), or cohort C (n=16). Three patients (one in cohort A and two in cohort B) were excluded due to ineligibility. 17 (22%) of 76 patients were female and 59 (78%) were male. At data cutoff (March 26, 2024), the median follow-up was 7·6 years (IQR 6·9-8·2). Overall intracranial responses occurred in 18 (51% [95% CI 34-69]) patients from cohort A, five (20% [7-41]) from cohort B, and one (6% [0-30]) from cohort C. 7-year intracranial progression-free survival was 42% (95% CI 29-63) in cohort A, 15% (6-39) in cohort B, and 6% (1-42) in cohort C. 7-year overall survival was 48% (34-68) in cohort A, 26% (13-51) in cohort B, and 13% (3-46) in cohort C. Safety results were consistent with the primary analysis. 50 patients died, including 18 (51%) from cohort A, 18 (72%) from cohort B, and 14 (88%) from cohort C. INTERPRETATION Our findings suggest that ipilimumab plus nivolumab maintains efficacy to at least 7 years in patients with active asymptomatic brain metastasis. Upfront ipilimumab plus nivolumab should be the standard of care for patients with melanoma brain metastasis; a trial investigating the role of stereotactic surgery in this new paradigm is ongoing. FUNDING Melanoma Institute Australia and Bristol Myers Squibb.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia.
| | - Victoria Atkinson
- Princess Alexandra Hospital, University of Queensland, Brisbane, QLD, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
| | - Alexander D Guminski
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia
| | - Shahneen K Sandhu
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Michael P Brown
- Royal Adelaide Hospital, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia; University of Adelaide, Adelaide, SA, Australia
| | - Maria Gonzalez
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
| | - Grant A McArthur
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia; Medical, Dental, and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia
| |
Collapse
|
7
|
Kooi EJ, Marcelis L, Wesseling P. Pathological diagnosis of central nervous system tumours in adults: what's new? Pathology 2025; 57:144-156. [PMID: 39818455 DOI: 10.1016/j.pathol.2024.11.004] [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: 10/10/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 01/18/2025]
Abstract
In the course of the last decade, the pathological diagnosis of many tumours of the central nervous system (CNS) has transitioned from a purely histological to a combined histological and molecular approach, resulting in a more precise 'histomolecular diagnosis'. Unfortunately, translation of this refinement in CNS tumour diagnostics into more effective treatment strategies is lagging behind. There is hope though that incorporating the assessment of predictive markers in the pathological evaluation of CNS tumours will help to improve this situation. The present review discusses some novel aspects with regard to the pathological diagnosis of the most common CNS tumours in adults. After a brief update on recognition of clinically meaningful subgroups in adult-type diffuse gliomas and the value of assessing predictive markers in these tumours, more detailed information is provided on predictive markers of (potential) relevance for immunotherapy especially for glioblastomas, IDH-wildtype. Furthermore, recommendations for improved grading of meningiomas by using molecular markers are briefly summarised, and an overview is given on (predictive) markers of interest in metastatic CNS tumours. In the last part of this review, some 'emerging new CNS tumour types' that may occur especially in adults are presented in a table. Hopefully, this review provides useful information on 'what's new' for practising pathologists diagnosing CNS tumours in adults.
Collapse
Affiliation(s)
- Evert-Jan Kooi
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Amsterdam, The Netherlands.
| | - Lukas Marcelis
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Amsterdam, The Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
8
|
Walsh N, Stephens R, Tan A, Durandt V, McLachlan J, Jordan J, Gregory K, Sutton S, Barrow C, Wong ANM. Real-World Outcomes of Immunotherapy for Melanoma Brain Metastases in New Zealand. JCO Oncol Pract 2025; 21:358-364. [PMID: 39052949 DOI: 10.1200/op.24.00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/08/2024] [Accepted: 04/21/2024] [Indexed: 07/27/2024] Open
Abstract
PURPOSE Melanoma brain metastases (BMs) are associated with poor survival. Combination immune checkpoint inhibitors (ICIs) with anti-PD1 and anti-CTLA-4 are the international standard-of-care treatment. Most landmark clinical trials excluded real-world patients with symptomatic disease, poor performance status (PS), and steroid use. Despite the high incidence of melanoma in New Zealand (NZ), the only publicly funded systemic treatment is anti-PD1 monotherapy. The real-world outcomes for BMs after ICIs in NZ are unknown. METHODOLOGY Medical records of patients with melanoma BMs in seven cancer centers across NZ between September 1, 2016, and September 1, 2020, were evaluated. Clinicopathologic characteristics, treatment, intracranial (IC) tumor response rates, IC progression-free survival, and overall survival (OS) are reported. RESULTS One hundred and forty-four patients received at least one dose of ICI. One hundred and thirty-three (93%) patients received anti-PD1 monotherapy. Almost a quarter of patients had poor baseline PS, 56% were symptomatic, and 33% had corticosteroids. Patients also received local therapies: 61 (42%) patients underwent surgery, 42 (29%) received whole brain radiation, and 47 (33%) received stereotactic radiation. The median OS was 15 months, and a third of patients were alive at 2 years. The toxicity of ICIs was at 28% and 15% for Common Terminology Criteria for Adverse Events grade 1-2 and 3-4 events, respectively. Of the patients who are still alive, 76% of patients remained symptomatic neurologically at last follow-up. CONCLUSION Most patients in this NZ real-world study were symptomatic and received anti-PD1 monotherapy. Approximately one-third of treated patients are alive at 2 years, but most patients remained symptomatic. This highlights the need for more effective treatment and prospective management of their neurologic rehabilitation needs.
Collapse
Affiliation(s)
| | | | - Alvin Tan
- Health New Zealand, New Zealand (National institution)
| | | | | | - Jody Jordan
- Health New Zealand, New Zealand (National institution)
| | - Kate Gregory
- Health New Zealand, New Zealand (National institution)
| | - Sean Sutton
- Health New Zealand, New Zealand (National institution)
| | | | - Annie N M Wong
- Wellington Blood & Cancer Centre, Wellington, New Zealand
| |
Collapse
|
9
|
Fischer GM, Lamba N, Vogelzang J, Aizer A, Ligon KL. Genomic Profiling Reveals SMARCA4 Mutations Are Associated with Shorter Overall and Intracranial Progression-Free Survival in Patients with Melanoma Brain Metastases. Clin Cancer Res 2025; 31:719-732. [PMID: 39786469 DOI: 10.1158/1078-0432.ccr-24-0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/12/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Melanoma brain metastases (MBM) are a common and lethal complication of metastatic melanoma. Despite improvements in treatments, subsets of patients with MBM experience rapid clinical decline, and currently, few prognostic biomarkers have been identified. An improved understanding of the molecular features specifically associated with MBM overall survival (OS) and intracranial progression-free survival (PFS) could facilitate the development of more effective clinical management strategies. EXPERIMENTAL DESIGN We established an initial cohort of 102 MBMs, 970 unmatched melanoma extracranial metastases (ECM), and 569 unmatched melanoma primaries with available targeted exome sequencing data covering 182 genes and a validation cohort of 50 MBMs with SMARCA4 genomically profiled. Kaplan-Meier analysis, log-rank test, and Cox proportional hazards model were used to evaluate associations between pathogenic genomic alterations and OS and intracranial PFS. We evaluated 14 MBMs and 19 ECMs with paired RNA sequencing and whole-exome sequencing data to identify genotype-transcriptome correlations. RESULTS Of 43 genes significantly mutated among MBMs, only pathogenic mutations in SMARCA4 significantly associated with shorter OS and intracranial PFS on univariable and multivariable analyses in patients with MBM but not from first ECM or primary tumor diagnosis. SMARCA4 mutations significantly associated with enrichment of oxidative phosphorylation and depletion of immune signaling gene sets. CONCLUSIONS Pathogenic SMARCA4 mutations independently predict an association with shorter OS and intracranial PFS in patients with MBM and associate with expression of pathways known to mediate melanoma virulence. These findings add to our understanding of MBM pathogenesis and suggest their potential use as prognostic biomarkers in patients with MBM and possible therapeutic opportunities.
Collapse
Affiliation(s)
- Grant M Fischer
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nayan Lamba
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jayne Vogelzang
- Department of Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ayal Aizer
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
10
|
Wikerholmen T, Taule EM, Rigg E, Berle BF, Sættem M, Sarnow K, Saed HS, Sundstrøm T, Thorsen F. Repurposing neuroleptics: clozapine as a novel, adjuvant therapy for melanoma brain metastases. Clin Exp Metastasis 2025; 42:12. [PMID: 39856383 PMCID: PMC11761981 DOI: 10.1007/s10585-025-10328-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/01/2025] [Indexed: 01/27/2025]
Abstract
The blood-brain barrier and the distinct brain immunology provide challenges in translating commonly used chemotherapeutics to treat intracranial tumors. Previous reports suggest anti-tumoral effects of antipsychotics, encouraging investigations into potential treatment effects of neuroleptics on brain metastases. For the first time, the therapeutic potential of the antipsychotic drug clozapine in treating melanoma brain metastases (MBM) was investigated using three human MBM cell lines. Through in vitro cell culture and viability experiments, clozapine displayed potent anti-tumoral effects on MBM cells with an exploitable therapeutic window when compared to normal human astrocytes or rat brain organoids. Further, it was shown that clozapine inhibited migration, proliferation, and colony formation in a dose-dependent manner. Through flow cytometry and proteome screening, we found that clozapine induced apoptosis in MBM cells and potentially altered the tumor immunological environment by upregulating proteins such as macrophage inflammatory protein-1 alpha (MIP-1α) and interleukin-8 (IL-8). In conclusion, clozapine shows significant and selective anti-tumoral effects on MBM cell lines in vitro. Further in vivo experiments are warranted to translate these results into clinical use.
Collapse
Affiliation(s)
- Tobias Wikerholmen
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Erlend Moen Taule
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Emma Rigg
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Birgitte Feginn Berle
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Magnus Sættem
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Katharina Sarnow
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
- Department of Neurosurgery, Boston Children's Hospital, 300 longwood Ave, Boston, MA, 02115, USA
| | - Halala Sdik Saed
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Terje Sundstrøm
- Department of Neurosurgery, Haukeland University Hospital, Haukelandsveien 22, Bergen, 5021, Norway
- Department of Clinical Medicine, University of Bergen, Jonas Lies Vei 87, Bergen, 5009, Norway
| | - Frits Thorsen
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway.
- Department of Neurosurgery, Haukeland University Hospital, Haukelandsveien 22, Bergen, 5021, Norway.
- Department of Clinical Medicine, University of Bergen, Jonas Lies Vei 87, Bergen, 5009, Norway.
- Molecular Imaging Center, Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway.
| |
Collapse
|
11
|
Guerrieri RA, Fischer GM, Kircher DA, Joon AY, Cortez JR, Grossman AH, Hudgens CW, Ledesma DA, Lazcano R, Onana CY, Knighton BG, Kumar S, Hu Q, Gopal YNV, McQuade JL, Deng W, Haydu LE, Gershenwald JE, Lazar AJ, Tetzlaff MT, Holmen SL, Davies MA. Oxidative Phosphorylation (OXPHOS) Promotes the Formation and Growth of Melanoma Lung and Brain Metastases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.633049. [PMID: 39896644 PMCID: PMC11785201 DOI: 10.1101/2025.01.23.633049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Melanoma mortality is driven by the formation and growth of distant metastases. Here, we interrogated the role of tumor oxidative phosphorylation (OXPHOS) in the formation of distant metastases in melanoma. OXPHOS was the most upregulated metabolic pathway in primary tumors that formed distant metastases in the RCAS-TVA mouse model of spontaneous lung and brain metastases, and in melanoma patients that developed brain or other distant metastases. Knockout of PGC1α in melanocytes in the RCAS-TVA melanoma mouse model had no impact on primary tumor formation, but markedly reduced the incidence of lung and brain metastases. Genetic knockout of a component of electron transport chain complex I, NDUFS4, in B16-F10 and D4M-UV2 murine melanoma cell lines did not impact tumor incidence following subcutaneous, intravenous, or intracranial injection, but decreased tumor burden specifically in the lungs and brain. Together, these data demonstrate that OXPHOS is critical for the formation of metastases in melanoma. STRUCTURED ABSTRACT Purpose: Melanoma mortality is driven by the formation and growth of distant metastases. However, the process and pathogenesis of melanoma metastasis remain poorly understood. Here, we interrogate the role of tumor oxidative phosphorylation (OXPHOS) in the formation of distant metastases in melanoma.Experimental Design: This study includes (1) new RNA-seq analysis of primary melanomas from patients characterized for distant metastasis events; (2) RNA-seq analysis and functional testing of genetic OXPHOS inhibition (PGC1α KO) the RCAS-TVA model, which is the only existing immunocompetent murine model of autochthonous lung and brain metastasis formation from primary melanoma tumors; and (3) functional experiments of genetic OXPHOS inhibition (NDUFS4 KO) in the B16-F10 and D4M-UV2 murine melanoma cell lines, including evaluation of subcutaneous, lung, and brain metastatic site dependencies.Results: OXPHOS was the most upregulated metabolic pathway in primary tumors that formed distant metastases in the RCAS-TVA mouse model of spontaneous lung and brain metastases, and in melanoma patients that developed brain or other distant metastases. Knockout of PGC1a in melanocytes in the RCAS-TVA melanoma mouse model had no impact on primary tumor formation, but markedly reduced the incidence of lung and brain metastases. Genetic knockout of a component of electron transport chain complex I, NDUFS4, in B16-F10 and D4M-UV2 murine melanoma cell lines did not impact tumor incidence following subcutaneous, intravenous, or intracranial injection, but decreased tumor burden specifically in the lungs and brain.Conclusions: Together, these data demonstrate that OXPHOS is critical for the formation of metastases in melanoma. TRANSLATIONAL RELEVANCE Melanoma is the most aggressive form of skin cancer. One hallmark of this disease is a high risk of distant metastasis formation. The process and pathogenesis of metastasis in this disease remain poorly understood and there is controversy regarding the role of oxidative phosphorylation (OXPHOS) in melanoma metastasis. This study incorporates RNAseq analysis of primary melanoma tumors from patients characterized for distant metastasis events, RNAseq analysis of the only existing immunocompetent murine model of autochthonous lung and brain metastasis formation from primary melanoma tumors, and functional testing in multiple syngeneic models of melanoma at different tissue sites. This integrated analysis consistently demonstrates that melanoma OXPHOS promotes distant metastasis to the lungs and brain, two of the most common and clinically relevant sites of melanoma metastasis. This improved understanding of tumor OXPHOS may represent novel vulnerabilities for therapeutics development and surveillance/preventative strategies for melanoma metastasis.
Collapse
|
12
|
Priego N, de Pablos-Aragoneses A, Perea-García M, Pieri V, Hernández-Oliver C, Álvaro-Espinosa L, Rojas A, Sánchez O, Steindl A, Caleiras E, García F, García-Martín S, Graña-Castro O, García-Mulero S, Serrano D, Velasco-Beltrán P, Jiménez-Lasheras B, Egia-Mendikute L, Rupp L, Stammberger A, Meinhardt M, Chaachou-Charradi A, Martínez-Saez E, Bertero L, Cassoni P, Mangherini L, Pellerino A, Rudà R, Soffietti R, Al-Shahrour F, Saftig P, Sanz-Pamplona R, Schmitz M, Crocker SJ, Calvo A, Palazón A, Valiente M. TIMP1 Mediates Astrocyte-Dependent Local Immunosuppression in Brain Metastasis Acting on Infiltrating CD8+ T Cells. Cancer Discov 2025; 15:179-201. [PMID: 39354883 PMCID: PMC11726018 DOI: 10.1158/2159-8290.cd-24-0134] [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: 01/31/2024] [Revised: 06/24/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024]
Abstract
Immunotherapies against brain metastases have shown clinical benefits when applied to asymptomatic patients, but they are largely ineffective in symptomatic cases for unknown reasons. Here, we dissect the heterogeneity in metastasis-associated astrocytes using single-cell RNA sequencing and report a population that blocks the antitumoral activity of infiltrating T cells. This protumoral activity is mediated by the secretion of tissue inhibitor of metalloproteinase-1 (TIMP1) from a cluster of pSTAT3+ astrocytes that acts on CD63+ CD8+ T cells to modulate their function. Using genetic and pharmacologic approaches in mouse and human brain metastasis models, we demonstrate that combining immune checkpoint blockade antibodies with the inhibition of astrocyte-mediated local immunosuppression may benefit patients with symptomatic brain metastases. We further reveal that the presence of tissue inhibitor of metalloproteinase-1 in liquid biopsies provides a biomarker to select patients for this combined immunotherapy. Overall, our findings demonstrate an unexpected immunomodulatory role for astrocytes in brain metastases with clinical implications. Significance: This study presents a significant advancement in understanding immune modulation in brain tumors and offers new insights into the potential therapeutic interventions for brain metastases. See related commentary by Lorger and James, p. 11.
Collapse
Affiliation(s)
- Neibla Priego
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - María Perea-García
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Valentina Pieri
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Laura Álvaro-Espinosa
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Andrea Rojas
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Oliva Sánchez
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ariane Steindl
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fernando García
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Osvaldo Graña-Castro
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sandra García-Mulero
- Biomarkers and Susceptibility Unit, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Department of Pathology and Experimental Therapy, School of Medicine, University of Barcelona (UB), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Diego Serrano
- Department of Pathology, Anatomy and Physiology, Faculty of Medicine, Center for Applied Clinical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
- CIBERONC, Madrid, Spain
| | - Paloma Velasco-Beltrán
- Cancer Immunology and Immunotherapy Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Borja Jiménez-Lasheras
- Cancer Immunology and Immunotherapy Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Leire Egia-Mendikute
- Cancer Immunology and Immunotherapy Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Luise Rupp
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, TU Dresden, Dresden, Germany
| | - Antonia Stammberger
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, TU Dresden, Dresden, Germany
| | - Matthias Meinhardt
- Department of Pathology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | | | - Luca Bertero
- Department of Medical Sciences, University of Turin, Turin, Italy
- Pathology Unit, Department of Laboratory Medicine, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Turin, Turin, Italy
- Pathology Unit, Department of Laboratory Medicine, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Luca Mangherini
- Department of Medical Sciences, University of Turin, Turin, Italy
- Pathology Unit, Department of Laboratory Medicine, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Alessia Pellerino
- Division of Neuro-Oncology, Department of Neuroscience, “Rita Levi Montalcini”, University and City of Health and Science Hospital, Turin, Italy
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, “Rita Levi Montalcini”, University and City of Health and Science Hospital, Turin, Italy
| | | | - Fatima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Paul Saftig
- Biochemical Institute, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Rebeca Sanz-Pamplona
- Biomarkers and Susceptibility Unit, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- University Hospital Lozano Blesa, Aragon Health Research Institute (IISA), ARAID Foundation, Aragon Government, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marc Schmitz
- Cancer Immunology and Immunotherapy Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephen J. Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Alfonso Calvo
- Department of Pathology, Anatomy and Physiology, Faculty of Medicine, Center for Applied Clinical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Asís Palazón
- CIBERONC, Madrid, Spain
- Ikerbasque, Basque Foundation for Science, Bizkaia, Spain
| | - RENACER
- Biobank, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
13
|
Podder V, Bellur S, Margolin K, Advani P, Mahtani RL, Subbiah V, Novo GB, Ranjan T, Ahluwalia MS. Immunotherapeutic and Targeted Strategies for Managing Brain Metastases from Common Cancer Origins: A State-of-the-Art Review. Curr Oncol Rep 2024; 26:1612-1638. [PMID: 39514054 DOI: 10.1007/s11912-024-01593-8] [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] [Accepted: 08/01/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW This review examines contemporary strategies for managing brain metastases (BM) from common cancers such as lung, breast, and melanoma. We evaluate the efficacy and applicability of targeted therapies and immunotherapies, exploring their potential to cross the blood-brain barrier and improve patient outcomes. RECENT FINDINGS Recent studies have shown that tyrosine kinase inhibitors, immune checkpoint inhibitors, and ADCs effectively treat BM. These treatments can overcome the challenges posed by the blood-brain barrier and improve therapeutic outcomes. ADCs are promising because they can deliver cytotoxic agents directly to tumor cells, which reduces systemic toxicity and increases drug delivery efficiency to the brain. Personalized medicine is becoming increasingly significant in treatment decisions, with biomarkers playing an essential role. Advances in molecular genetics and drug development have led to more refined treatments, emphasizing the precision medicine framework. The management of BM is evolving, driven by drug efficacy, resistance mechanisms, and the need for personalized medicine. Integrating ADCs into treatment regimens represents a significant advancement in targeting metastatic brain tumors. Despite these advances, BM management still presents considerable challenges, requiring ongoing research and multi-institutional trials to optimize therapeutic strategies. This review outlines the current state and future directions in treating BM, highlighting the critical need for continued innovation and comprehensive clinical evaluations to improve survival rates and quality of life for affected patients.
Collapse
Affiliation(s)
- Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Shreyas Bellur
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Kim Margolin
- Saint John's Cancer Institute, Santa Monica, CA, USA
| | | | - Reshma L Mahtani
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Vivek Subbiah
- Sarah Cannon Research Institute (SCRI), Nashville, TN, USA
| | - Gabriella B Novo
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Tulika Ranjan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | |
Collapse
|
14
|
Alhusaini S, Naya L, Reddy SA, Patel CB. MEK Pathway Inhibitor-Mediated Response in BRAF V600-Mutant Melanoma with Brain Parenchymal and Leptomeningeal Metastases. Ann Neurol 2024; 96:1227-1229. [PMID: 39324488 DOI: 10.1002/ana.27062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/27/2024]
Affiliation(s)
- Saud Alhusaini
- Department of Neurology, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Lewis Naya
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sunil A Reddy
- Division of Melanoma Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Chirag B Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
15
|
Samoylenko IV, Kolontareva YM, Kogay EV, Zhukova NV, Utyashev IA, Ivannikov ME, Menshikov KV, Zinkevich MV, Orlova KV, Vakhabova YV, Volkonsky MV, Beliaeva NA, Butkov II, Karabina EV, Moskovkina TL, Moshkova KA, Plishkina OV, Sychev VD, Cheplukhova OS, Chernova VV, Yurchenkov AN, Babina KG, Savelov NA, Demidov LV. Triple combination of vemurafenib, cobimetinib, and atezolizumab in real clinical practice in the Russian Federation: results of the A1 cohort of the ISABELLA study. Front Oncol 2024; 14:1395378. [PMID: 39469641 PMCID: PMC11514068 DOI: 10.3389/fonc.2024.1395378] [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: 03/03/2024] [Accepted: 08/26/2024] [Indexed: 10/30/2024] Open
Abstract
Background Among several treatment options for BRAF-mutant metastatic melanoma, a combination of BRAF inhibitor, MEK inhibitor, and anti-PDL1 antibody seems to be a new emergent approach recently registered in the Russian Federation. It is still not clear which patient population benefits more from this simultaneous use of three drugs instead of its sequencing. Aim This study aimed to evaluate patients' characteristics treated in real practice in 14 Russian regions by triple combination and to analyze their outcomes depending on biomarkers (PD-L1 expression). Methods This was a part (cohort A1) of a prospective non-interventional study of clinical outcomes and biomarkers in patients with skin melanoma. Patients were included in cohort A1 if combination treatment with vemurafenib (vem) + cobimetinib (cobi) + atezolizumab (atezo) was initiated no earlier than 12 weeks (84 days) prior to written informed consent to participate in this study. The index event was the initiation of therapy with all three drugs vem + cobi + atezo (i.e., triple combination). The primary efficacy endpoint of the study was the 24-month overall survival (OS), defined as the time from the index date to the date of death from any cause. If the patient did not experience an event, the OS will be censored at the date of the last contact. Objective response rate (ORR), duration of response (DoR), and progression-free survival (PFS) in the Intention to treat (ITT) population, in biomarker positive population, and in population with brain metastases were also evaluated. Quality of life questionnaires were pre-planned by protocol if it was a part of routine practice. Adverse events were also collected. Results Between March 2021 and May 2023, 59 patients were enrolled in 19 centers from 14 regions of Russia. Thirty-one of 59 (52.4%) patients had central nervous system metastases, and 18 of 31 (58.4%) were symptomatic. Forty of 59 patients (68%) received the triple combination as the first-line treatment. The median follow-up period was 16.83 [95% confidence interval (CI) 13.8-19.8] months. The mean duration of therapy with this regimen was 9.95 months (95% CI 7.48-13.8). ORR was 55.1%; progression as the best outcome was seen in 16.3%. The median DoR was 12.95 months (95% CI 11.0-14.8 months), with a median of 20.3 months (95% CI 9.1-31.5 months) when triple therapy was administered in the first-line treatment. In patients with brain metastases (N = 31), ORR was 45.1%; the median DoR was 12.95 (95% CI 11.0-14.8 months). The median PFS in the entire population was 13.6 months (95% CI 8.6-18.6); the 24-month PFS was 22%. The estimated median OS in the entire population was 15.8 months (95% CI NA); 24-month OS was 45% (95% CI 0.32-0.64). In multivariate Cox regression model, biomarkers of interest [lactate dehydrogenase, Programmed cell death ligand-1 (PD-L1)] did not have statistically significant impact on PFS, OS, or DoR probably due to high data missing rate. No unexpected adverse events were reported. Grades 3-4 AEs were seen in 23 of 59 patients (38%) with most common were skin and liver toxicity. Conclusion Triple combination of atezolizumab, vemurafenib, and cobimetinib had proven its efficacy and tolerability in real settings. No impact of potential predictive biomarkers was seen (NCT05402059).
Collapse
Affiliation(s)
- Igor V. Samoylenko
- Skin tumors department, NN Blokhin National Medical Research Center of Oncology, Moscow, Russia
- The Russian Melanoma Professional Association (Melanoma.PRO), Moscow, Russia
| | | | - Ekaterina V. Kogay
- Skin tumors department, NN Blokhin National Medical Research Center of Oncology, Moscow, Russia
- The Russian Melanoma Professional Association (Melanoma.PRO), Moscow, Russia
| | - Natalia V. Zhukova
- St. Petersburg City Clinical Oncology Dispensary, St. Petersburg, Russia
| | | | | | - Konstantin V. Menshikov
- Ufa Republican Clinical Oncological Dispensary of the Ministry of Health Republic of Bashkortostan, Ufa, Russia
| | - Maxim V. Zinkevich
- Leningrad Regional Clinical Oncological Dispensary, St. Petersburg, Russia
| | - Kristina V. Orlova
- Skin tumors department, NN Blokhin National Medical Research Center of Oncology, Moscow, Russia
- The Russian Melanoma Professional Association (Melanoma.PRO), Moscow, Russia
| | | | | | | | | | | | | | - Kseniya A. Moshkova
- Nizhny Novgorod Regional Clinical Oncological Dispensary, Nizhny Novgorod, Russia
| | | | | | | | | | | | - Ksenia G. Babina
- Volgograd Regional Clinical Oncological Dispensary, Volgograd, Russia
| | | | - Lev V. Demidov
- Skin tumors department, NN Blokhin National Medical Research Center of Oncology, Moscow, Russia
- The Russian Melanoma Professional Association (Melanoma.PRO), Moscow, Russia
| |
Collapse
|
16
|
Daugherty-Lopès A, Pérez-Guijarro E, Gopalan V, Rappaport J, Chen Q, Huang A, Lam KC, Chin S, Ebersole J, Wu E, Needle GA, Church I, Kyriakopoulos G, Xie S, Zhao Y, Gruen C, Sassano A, Araya RE, Thorkelsson A, Smith C, Lee MP, Hannenhalli S, Day CP, Merlino G, Goldszmid RS. IMMUNE AND MOLECULAR CORRELATES OF RESPONSE TO IMMUNOTHERAPY REVEALED BY BRAIN-METASTATIC MELANOMA MODELS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609785. [PMID: 39372744 PMCID: PMC11451731 DOI: 10.1101/2024.08.26.609785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Despite the promising results of immune checkpoint blockade (ICB) therapy, outcomes for patients with brain metastasis (BrM) remain poor. Identifying resistance mechanisms has been hindered by limited access to patient samples and relevant preclinical models. Here, we developed two mouse melanoma BrM models that recapitulate the disparate responses to ICB seen in patients. We demonstrate that these models capture the cellular and molecular complexity of human disease and reveal key factors shaping the tumor microenvironment and influencing ICB response. BR1-responsive tumor cells express inflammatory programs that polarize microglia into reactive states, eliciting robust T cell recruitment. In contrast, BR3-resistant melanoma cells are enriched in neurological programs and exploit tolerance mechanisms to maintain microglia homeostasis and limit T cell infiltration. In humans, BR1 and BR3 expression signatures correlate positively or negatively with T cell infiltration and BrM patient outcomes, respectively. Our study provides clinically relevant models and uncovers mechanistic insights into BrM ICB responses, offering potential biomarkers and therapeutic targets to improve therapy efficacy.
Collapse
Affiliation(s)
- Amélie Daugherty-Lopès
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Eva Pérez-Guijarro
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jessica Rappaport
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Quanyi Chen
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Kelly Government Solutions, Bethesda, MD, USA
| | - April Huang
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Kelly Government Solutions, Bethesda, MD, USA
| | - Khiem C. Lam
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sung Chin
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Jessica Ebersole
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Emily Wu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Gabriel A. Needle
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Isabella Church
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - George Kyriakopoulos
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Shaojun Xie
- CCR-SF Bioinformatics Team, Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Yongmei Zhao
- CCR-SF Bioinformatics Team, Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Charli Gruen
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Antonella Sassano
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Romina E. Araya
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Andres Thorkelsson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Cari Smith
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Maxwell P. Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Romina S. Goldszmid
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Hasanov M, Acikgoz Y, Davies MA. Melanoma Brain Metastasis: Biology and Therapeutic Advances. Hematol Oncol Clin North Am 2024; 38:1027-1043. [PMID: 38845301 DOI: 10.1016/j.hoc.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2024]
Abstract
Metastasis to the brain is a frequent complication of advanced melanoma. Historically, patients with melanoma brain metastasis (MBM) have had dismal outcomes, but outcomes have improved with the development of more effective treatments, including stereotactic radiosurgery and effective immune and targeted therapies. Despite these advances, MBM remains a leading cause of death from this disease, and many therapies show decreased efficacy against these tumors compared with extracranial metastases. This differential efficacy may be because of recently revealed unique molecular and immune features of MBMs-which may also provide rational new therapeutic strategies.
Collapse
Affiliation(s)
- Merve Hasanov
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Suite 1335, Lincoln Tower, 1800 Cannon Drive, Columbus, OH, 43210, USA.
| | - Yusuf Acikgoz
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 13th floor, Lincoln Tower, 1800 Cannon Drive, Columbus, OH, 43210, USA
| | - Michael A Davies
- Division of Cancer Medicine, Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0430, Houston, TX 77030, USA
| |
Collapse
|
18
|
Sloan AE, Winter K, Gilbert MR, Aldape K, Choi S, Wen PY, Butowski N, Iwamoto FM, Raval RR, Voloschin AD, Kamiya-Matsuoka C, Won M, Mehta MP. NRG-BN002: Phase I study of ipilimumab, nivolumab, and the combination in patients with newly diagnosed glioblastoma. Neuro Oncol 2024; 26:1628-1637. [PMID: 38874333 PMCID: PMC11376446 DOI: 10.1093/neuonc/noae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have efficacy in several solid tumors but limited efficacy in glioblastoma (GBM). This study evaluated the safety of anti-CTLA-4 and anti-PD-1 ICIs alone or in combination in newly diagnosed GBM after completion of standard radiochemotherapy with the subsequent intent to test combinatorial ICIs in this setting. METHODS The primary endpoint was dose-limiting toxicity (DLT) for adults with unifocal, supratentorial newly diagnosed GBM after resection and chemoradiation. Ipilimumab and nivolumab were tested separately and in combination with a planned expansion cohort dependent upon DLT results. RESULTS Thirty-two patients were enrolled at 9 institutions: 6 to each DLT assessment cohort and 14 to the expansion cohort. Median age: 55 years, 67.7% male, 83.9% White. Treatment was well tolerated with 16% Grade 4 events; the combination did not have unexpectedly increased toxicity, with no Grade 5 events. One DLT was seen in each single-agent treatment; none were observed in the combination, leading to expanded accrual of the combined treatment. The median follow-up was 19.6 months. For all patients receiving combination treatment, median overall survival (OS) and progression-free survival (PFS) were 20.7 and 16.1 months, respectively. CONCLUSIONS IPI and NIVO are safe and tolerable with toxicities similar to those noted with other cancers when given in combination with adjuvant temozolomide for newly diagnosed GBM. Combination IPI + NIVO is not substantially more toxic than single agents. These results support a subsequent efficacy trial to test the combination of ICIs in Phase II/III for patients with newly diagnosed GBM. CLINICALTRIALS.GOV REGISTRATION NCT02311920.
Collapse
Affiliation(s)
- Andrew E Sloan
- Piedmont Healthcare, Atlanta, Georgia, USA
- Case Western Reserve University & Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Kathryn Winter
- NRG Oncology Statistics and Data Management Center/ACR, Philadelphia, Pennsylvania, USA
- American College of Radiology, Philadelphia, Pennsylvania, USA
| | - Mark R Gilbert
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Kenneth Aldape
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Serah Choi
- Case Western Reserve University & Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Patrick Y Wen
- Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| | | | - Fabio M Iwamoto
- Columbia University Minority Underserved NCORP, New York, New York, USA
| | - Raju R Raval
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | | | | | - Minhee Won
- NRG Oncology Statistics and Data Management Center/ACR, Philadelphia, Pennsylvania, USA
- Case Western Reserve University & Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | | |
Collapse
|
19
|
Khan B, Qahwaji RM, Alfaifi MS, Mobashir M. Nivolumab and Ipilimumab Acting as Tormentors of Advanced Tumors by Unleashing Immune Cells and Associated Collateral Damage. Pharmaceutics 2024; 16:732. [PMID: 38931856 PMCID: PMC11207028 DOI: 10.3390/pharmaceutics16060732] [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: 03/18/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 06/28/2024] Open
Abstract
Combining immune checkpoint inhibitors, specifically nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4), holds substantial promise in revolutionizing cancer treatment. This review explores the transformative impact of these combinations, emphasizing their potential for enhancing therapeutic outcomes across various cancers. Immune checkpoint proteins, such as PD1 and CTLA4, play a pivotal role in modulating immune responses. Blocking these checkpoints unleashes anticancer activity, and the synergy observed when combining multiple checkpoint inhibitors underscores their potential for enhanced efficacy. Nivolumab and ipilimumab harness the host's immune system to target cancer cells, presenting a powerful approach to prevent tumor development. Despite their efficacy, immune checkpoint inhibitors are accompanied by a distinct set of adverse effects, particularly immune-related adverse effects affecting various organs. Understanding these challenges is crucial for optimizing treatment strategies and ensuring patient well-being. Ongoing clinical trials are actively exploring the combination of checkpoint inhibitory therapies, aiming to decipher their synergistic effects and efficacy against diverse cancer types. This review discusses the mechanisms, adverse effects, and various clinical trials involving nivolumab and ipilimumab across different cancers, emphasizing their transformative impact on cancer treatment.
Collapse
Affiliation(s)
- Bushra Khan
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Rowaid M. Qahwaji
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22233, Saudi Arabia;
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashael S. Alfaifi
- Department of Epidemiology, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Mohammad Mobashir
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Solnavägen 9, 171 65 Solna, Sweden
| |
Collapse
|
20
|
Kraft T, Grützmann K, Meinhardt M, Meier F, Westphal D, Seifert M. Personalized identification and characterization of genome-wide gene expression differences between patient-matched intracranial and extracranial melanoma metastasis pairs. Acta Neuropathol Commun 2024; 12:67. [PMID: 38671536 PMCID: PMC11055243 DOI: 10.1186/s40478-024-01764-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Melanoma is the most serious type of skin cancer that frequently spreads to other organs of the human body. Especially melanoma metastases to the brain (intracranial metastases) are hard to treat and a major cause of death of melanoma patients. Little is known about molecular alterations and altered mechanisms that distinguish intra- from extracranial melanoma metastases. So far, almost all existing studies compared intracranial metastases from one set of patients to extracranial metastases of an another set of melanoma patients. This neglects the important facts that each melanoma is highly individual and that intra- and extracranial melanoma metastases from the same patient are more similar to each other than to melanoma metastases from other patients in the same organ. To overcome this, we compared the gene expression profiles of 16 intracranial metastases to their corresponding 21 patient-matched extracranial metastases in a personalized way using a three-state Hidden Markov Model (HMM) to identify altered genes for each individual metastasis pair. This enabled three major findings by considering the predicted gene expression alterations across all patients: (i) most frequently altered pathways include cytokine-receptor interaction, calcium signaling, ECM-receptor interaction, cAMP signaling, Jak-STAT and PI3K/Akt signaling, (ii) immune-relevant signaling pathway genes were downregulated in intracranial metastases, and (iii) intracranial metastases were associated with a brain-like phenotype gene expression program. Further, the integration of all differentially expressed genes across the patient-matched melanoma metastasis pairs led to a set of 103 genes that were consistently down- or up-regulated in at least 11 of the 16 of the patients. This set of genes contained many genes involved in the regulation of immune responses, cell growth, cellular signaling and transport processes. An analysis of these genes in the TCGA melanoma cohort showed that the expression behavior of 11 genes was significantly associated with survival. Moreover, a comparison of the 103 genes to three closely related melanoma metastasis studies revealed a core set of eight genes that were consistently down- or upregulated in intra- compared to extracranial metastases in at least two of the three related studies (down: CILP, DPT, FGF7, LAMP3, MEOX2, TMEM119; up: GLDN, PMP2) including FGF7 that was also significantly associated with survival. Our findings contribute to a better characterization of genes and pathways that distinguish intra- from extracranial melanoma metastasis and provide important hints for future experimental studies to identify potential targets for new therapeutic approaches.
Collapse
Affiliation(s)
- Theresa Kraft
- Institute for Medical Informatics and Biometry (IMB), Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Konrad Grützmann
- Institute for Medical Informatics and Biometry (IMB), Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Matthias Meinhardt
- Department of Pathology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- Skin Cancer Center at the University Cancer Center (UCC) Dresden and the National Center for Tumor Diseases Dresden (NCT), Fetscherstr. 74, 01307, Dresden, Germany
| | - Dana Westphal
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT), Fetscherstr. 74, 01307, Dresden, Germany
| | - Michael Seifert
- Institute for Medical Informatics and Biometry (IMB), Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- National Center for Tumor Diseases Dresden (NCT), Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
21
|
Boutros A, Croce E, Ferrari M, Gili R, Massaro G, Marconcini R, Arecco L, Tanda ET, Spagnolo F. The treatment of advanced melanoma: Current approaches and new challenges. Crit Rev Oncol Hematol 2024; 196:104276. [PMID: 38295889 DOI: 10.1016/j.critrevonc.2024.104276] [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: 09/30/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/17/2024] Open
Abstract
In recent years, advances in melanoma treatment have renewed patient hope. This comprehensive review emphasizes the evolving treatment landscape, particularly highlighting first-line strategies and the interplay between immune-checkpoint inhibitors (ICIs) and targeted therapies. Ipilimumab plus nivolumab has achieved the best median overall survival, exceeding 70 months. However, the introduction of new ICIs, like relatlimab, has added complexity to first-line therapy decisions. Our aim is to guide clinicians in making personalized treatment decisions. Various features, including brain metastases, PD-L1 expression, BRAF mutation, performance status, and prior adjuvant therapy, significantly impact the direction of advanced melanoma treatment. We also provide the latest insights into the treatment of rare melanoma subtypes, such as uveal melanoma, where tebentafusp has shown promising improvements in overall survival for metastatic uveal melanoma patients. This review provides invaluable insights for clinicians, enabling informed treatment choices and deepening our understanding of the multifaceted challenges associated with advanced melanoma management.
Collapse
Affiliation(s)
- Andrea Boutros
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy.
| | - Elena Croce
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Ferrari
- Azienda Ospedaliero Universitaria Pisana, Medical Oncology Unit, Pisa, Italy
| | - Riccardo Gili
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Giulia Massaro
- Unit of Medical Oncology, Careggi University-Hospital, 50134 Florence, Italy
| | - Riccardo Marconcini
- Azienda Ospedaliero Universitaria Pisana, Medical Oncology Unit, Pisa, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy; Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Enrica Teresa Tanda
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Spagnolo
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genova, Italy
| |
Collapse
|
22
|
Knox A, Wang T, Shackleton M, Ameratunga M. Symptomatic brain metastases in melanoma. Exp Dermatol 2024; 33:e15075. [PMID: 38610093 DOI: 10.1111/exd.15075] [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: 01/11/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024]
Abstract
Although clinical outcomes in metastatic melanoma have improved in recent years, the morbidity and mortality of symptomatic brain metastases remain challenging. Response rates and survival outcomes of patients with symptomatic melanoma brain metastases (MBM) are significantly inferior to patients with asymptomatic disease. This review focusses upon the specific challenges associated with the management of symptomatic MBM, discussing current treatment paradigms, obstacles to improving clinical outcomes and directions for future research.
Collapse
Affiliation(s)
- Andrea Knox
- Department of Medical Oncology, Alfred Health, Melbourne, Australia
| | - Tim Wang
- Department of Radiation Oncology, Westmead Hospital, Sydney, Australia
| | - Mark Shackleton
- Department of Medical Oncology, Alfred Health, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Malaka Ameratunga
- Department of Medical Oncology, Alfred Health, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
23
|
Namikawa K, Nakano E, Ogata D, Yamazaki N. Long-term survival with systemic therapy in the last decade: Can melanoma be cured? J Dermatol 2024; 51:343-352. [PMID: 38358050 PMCID: PMC11484129 DOI: 10.1111/1346-8138.17147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Immune checkpoint inhibitors have been shown to prolong survival of patients with several types of cancer, and the finding was first established in melanoma. Previously, systemic therapy for advanced melanoma aimed only at tumor control and palliation of symptoms. However, in recent years, some patients who received systemic therapy have achieved a complete response and survived without continuous treatment for more than several years. This review discusses the long-term survival rates achieved with currently used systemic therapies and their future perspectives. Long-term survival is currently most likely to be achieved with the use of the standard-dose combination of nivolumab plus ipilimumab, however, this regimen is associated with a high frequency of serious or persistent immune-related adverse events. Several new anti-PD-1-based combination therapies with a better risk-benefit balance are currently under development. Although the acral and mucosal subtypes tend to be less responsive to immune checkpoint inhibitors, anti-PD-1-based combination therapy should continue to be investigated for these subtypes owing to its potential for better long-term survival. With the development of efficacious immunotherapy and targeted therapy, it is important to determine the optimal duration of systemic therapy to avoid unnecessary health and financial burdens as well as to improve efforts to support long-term cancer survivors. As the goal of systemic therapy shifts from tumor control to long-term survival, in future clinical trials, long-term clinical outcomes should be evaluated to assess the benefits of novel agents.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Eiji Nakano
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Dai Ogata
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Naoya Yamazaki
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
24
|
Nayyar N, de Sauvage MA, Chuprin J, Sullivan EM, Singh M, Torrini C, Zhang BS, Bandyopadhyay S, Daniels KA, Alvarez-Breckenridge C, Dahal A, Brehm MA, Brastianos PK. CDK4/6 Inhibition Sensitizes Intracranial Tumors to PD-1 Blockade in Preclinical Models of Brain Metastasis. Clin Cancer Res 2024; 30:420-435. [PMID: 37611074 PMCID: PMC10872577 DOI: 10.1158/1078-0432.ccr-23-0433] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/15/2023] [Accepted: 08/22/2023] [Indexed: 08/25/2023]
Abstract
PURPOSE Brain metastases are associated with high morbidity and are often resistant to immune checkpoint inhibitors. We evaluated whether CDK4/6 inhibitor (CDKi) abemaciclib can sensitize intracranial tumors to programmed cell death protein 1 (PD-1) inhibition in mouse models of melanoma and breast cancer brain metastasis. EXPERIMENTAL DESIGN Treatment response was evaluated in vivo using immunocompetent mouse models of brain metastasis bearing concurrent intracranial and extracranial tumors. Treatment effect on intracranial and extracranial tumor-immune microenvironments (TIME) was evaluated using immunofluorescence, multiplex immunoassays, high-parameter flow cytometry, and T-cell receptor profiling. Mice with humanized immune systems were evaluated using flow cytometry to study the effect of CDKi on human T-cell development. RESULTS We found that combining abemaciclib with PD-1 inhibition reduced tumor burden and improved overall survival in mice. The TIME, which differed on the basis of anatomic location of tumors, was altered with CDKi and PD-1 inhibition in an organ-specific manner. Combination abemaciclib and anti-PD-1 treatment increased recruitment and expansion of CD8+ effector T-cell subsets, depleted CD4+ regulatory T (Treg) cells, and reduced levels of immunosuppressive cytokines in intracranial tumors. In immunodeficient mice engrafted with human immune systems, abemaciclib treatment supported development and maintenance of CD8+ T cells and depleted Treg cells. CONCLUSIONS Our results highlight the distinct properties of intracranial and extracranial tumors and support clinical investigation of combination CDK4/6 and PD-1 inhibition in patients with brain metastases. See related commentary by Margolin, p. 257.
Collapse
Affiliation(s)
- Naema Nayyar
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| | | | - Jane Chuprin
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA
| | - Emily M Sullivan
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| | - Mohini Singh
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| | - Consuelo Torrini
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| | - Britney S Zhang
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| | - Sushobhana Bandyopadhyay
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania
| | - Keith A Daniels
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA
| | - Christopher Alvarez-Breckenridge
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ashish Dahal
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| | - Michael A Brehm
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| | - Priscilla K Brastianos
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
25
|
Meng L, Wu H, Wu J, Ding P, He J, Sang M, Liu L. Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer hallmarks. Cell Death Dis 2024; 15:3. [PMID: 38177102 PMCID: PMC10766988 DOI: 10.1038/s41419-023-06389-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 01/06/2024]
Abstract
Current treatment strategies for cancer, especially advanced cancer, are limited and unsatisfactory. One of the most substantial advances in cancer therapy, in the last decades, was the discovery of a new layer of immunotherapy approach, immune checkpoint inhibitors (ICIs), which can specifically activate immune cells by targeting immune checkpoints. Immune checkpoints are a type of immunosuppressive molecules expressed on immune cells, which can regulate the degree of immune activation and avoid autoimmune responses. ICIs, such as anti-PD-1/PD-L1 drugs, has shown inspiring efficacy and broad applicability across various cancers. Unfortunately, not all cancer patients benefit remarkably from ICIs, and the overall response rates to ICIs remain relatively low for most cancer types. Moreover, the primary and acquired resistance to ICIs pose serious challenges to the clinical application of cancer immunotherapy. Thus, a deeper understanding of the molecular biological properties and regulatory mechanisms of immune checkpoints is urgently needed to improve clinical options for current therapies. Recently, circular RNAs (circRNAs) have attracted increasing attention, not only due to their involvement in various aspects of cancer hallmarks, but also for their impact on immune checkpoints in shaping the tumor immune microenvironment. In this review, we systematically summarize the current status of immune checkpoints in cancer and the existing regulatory roles of circRNAs on immune checkpoints. Meanwhile, we also aim to settle the issue in an evidence-oriented manner that circRNAs involved in cancer hallmarks regulate the effects and resistance of ICIs by targeting immune checkpoints.
Collapse
Affiliation(s)
- Lingjiao Meng
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050035, China
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Haotian Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Jiaxiang Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Ping'an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Jinchen He
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Meixiang Sang
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
- Science and Education Department, Shanghai Electric Power Hospital, Shanghai, 20050, China.
| | - Lihua Liu
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050035, China.
| |
Collapse
|
26
|
Zhou D, Gong Z, Wu D, Ma C, Hou L, Niu X, Xu T. Harnessing immunotherapy for brain metastases: insights into tumor-brain microenvironment interactions and emerging treatment modalities. J Hematol Oncol 2023; 16:121. [PMID: 38104104 PMCID: PMC10725587 DOI: 10.1186/s13045-023-01518-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023] Open
Abstract
Brain metastases signify a deleterious milestone in the progression of several advanced cancers, predominantly originating from lung, breast and melanoma malignancies, with a median survival timeframe nearing six months. Existing therapeutic regimens yield suboptimal outcomes; however, burgeoning insights into the tumor microenvironment, particularly the immunosuppressive milieu engendered by tumor-brain interplay, posit immunotherapy as a promising avenue for ameliorating brain metastases. In this review, we meticulously delineate the research advancements concerning the microenvironment of brain metastases, striving to elucidate the panorama of their onset and evolution. We encapsulate three emergent immunotherapeutic strategies, namely immune checkpoint inhibition, chimeric antigen receptor (CAR) T cell transplantation and glial cell-targeted immunoenhancement. We underscore the imperative of aligning immunotherapy development with in-depth understanding of the tumor microenvironment and engendering innovative delivery platforms. Moreover, the integration with established or avant-garde physical methodologies and localized applications warrants consideration in the prevailing therapeutic schema.
Collapse
Affiliation(s)
- Dairan Zhou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Zhenyu Gong
- Department of Neurosurgery, Klinikum Rechts Der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Chao Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Lijun Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Xiaomin Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Xuhui District, Shanghai, 200030, People's Republic of China.
| | - Tao Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
27
|
Su M, Yang Y, Wang P. Efficacy and safety of the combined use of ipilimumab and nivolumab for melanoma patients with brain metastases: a systematic review and meta-analysis. Immunopharmacol Immunotoxicol 2023; 45:761-769. [PMID: 37228242 DOI: 10.1080/08923973.2023.2215403] [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: 03/15/2023] [Accepted: 05/14/2023] [Indexed: 05/27/2023]
Abstract
CONTEXT Immune checkpoint inhibitors have advanced immunotherapy for melanoma patients.Objective: This study evaluates efficacy and safety of ipilimumab and nivolumab combination (IN) for melanoma brain metastases (MBM) patients. MATERIALS AND METHODS Literature search was conducted in electronic databases and studies were included if they reported efficacy and safety of IN in MBM patients or prognostic information related to brain metastases. Outcomes evaluated were objective response rate (ORR), complete remission/stable disease/progressive disease rates, progression-free survival (PFS), overall survival (OS), incidence rates of adverse events, and hazard ratios of disease progression or mortality between IN-treated patients with and without brain metastasis. RESULTS Intracranial ORR was higher in IN-treated MBM patients than with control therapies (nivolumab or ipilimumab plus fotemustine). IN treatment led to longer PFS and OS in than control treatments. Five-year OS of IN-treated MBM patients was up to 51% compared to 34% for nivolumab. Outcomes were better for treatment naïve and asymptomatic patients. Whereas many studies reported significantly higher mortality or progression risk with IN treatment in MBM patients compared to non-MBM melanoma patients, many others did not find this risk significant. Incidence of grade 3/4 adverse events in IN-treated MBM patients was: diarrhea or colitis (16%), hepatitis (15%), rash (8%), increased alanine transaminase (8%), increased aspartate aminotransferase (7%), increased lipase (6%), increased amylase (4%), fatigue (3%), hypophysitis (2%), pneumonitis (2%), headache (2%), nausea or vomiting (1%), and neutropenia (1%). CONCLUSION IN is an efficacious and safer treatment option for MBM patients, especially for asymptomatic and treatment naïve patients.
Collapse
Affiliation(s)
- Mengmeng Su
- Department of Radiotherapy, Peking University People's Hospital, Beijing, China
| | - Yuyan Yang
- Department of Radiotherapy, Peking University International Hospital, Beijing, China
| | - Peng Wang
- Department of Radiotherapy, Peking University International Hospital, Beijing, China
| |
Collapse
|
28
|
Kumthekar P, Le Rhun E. Brain Metastases and Leptomeningeal Disease. Continuum (Minneap Minn) 2023; 29:1727-1751. [PMID: 38085896 DOI: 10.1212/con.0000000000001354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
OBJECTIVE Central nervous system (CNS) metastases include brain parenchymal, spinal cord, and leptomeningeal metastases. This article discusses the diagnostic and therapeutic advances of the last decade that have improved outcomes for patients with these CNS metastases. LATEST DEVELOPMENTS The diagnostic tools for CNS metastases, particularly leptomeningeal disease, have evolved over the past decade with respect to advancements in CSF analysis. Multiple medical therapies are now available for brain metastasis treatment that have shown CNS efficacy, including targeted therapies and antibody-drug conjugates. Molecular testing for CNS metastases has become more common and the repertoire of molecularly targeted therapies continues to expand. Advancements in radiation therapy, including improvements in stereotactic radiation techniques, whole-brain radiation with hippocampal avoidance, and proton beam radiation, have changed the radiation management of patients with CNS metastases. New intrathecal agents are currently being tested for the management of leptomeningeal metastases. ESSENTIAL POINTS CNS metastases are far more common than primary brain tumors and are increasing in prevalence in the setting of improved treatments and prolonged survival in patients with systemic cancers. There have been many changes in the diagnostics and treatment of CNS metastases, yielding subsequent improvements in patient outcomes with further advancements on the horizon.
Collapse
|
29
|
Dummer R, Queirolo P, Gerard Duhard P, Hu Y, Wang D, de Azevedo SJ, Robert C, Ascierto PA, Chiarion-Sileni V, Pronzato P, Spagnolo F, Mujika Eizmendi K, Liszkay G, de la Cruz Merino L, Tawbi H. Atezolizumab, vemurafenib, and cobimetinib in patients with melanoma with CNS metastases (TRICOTEL): a multicentre, open-label, single-arm, phase 2 study. Lancet Oncol 2023; 24:e461-e471. [PMID: 37459873 DOI: 10.1016/s1470-2045(23)00334-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
BACKGROUND Targeted therapy and immunotherapy have shown intracranial activity in melanoma with CNS metastases, but there remains an unmet need, particularly for patients with symptomatic CNS metastases. We aimed to evaluate atezolizumab in combination with cobimetinib or vemurafenib plus cobimetinib in patients with melanoma with CNS metastases. METHODS TRICOTEL was a multicentre, open-label, single-arm, phase 2 study done in two cohorts: a BRAFV600 wild-type cohort and a BRAFV600 mutation-positive cohort, recruited at 21 hospitals and oncology centres in Brazil, France, Germany, Hungary, Italy, Spain, and Switzerland. Eligible patients were aged 18 years or older with previously untreated metastatic melanoma, brain metastases of 5 mm or larger in at least one dimension, and an Eastern Cooperative Oncology Group performance status of 2 or less. Patients in the BRAFV600 wild-type cohort received intravenous atezolizumab (840 mg, days 1 and 15 of each 28-day cycle) plus oral cobimetinib (60 mg once daily, days 1-21). Patients in the BRAFV600 mutation-positive cohort received intravenous atezolizumab (840 mg, days 1 and 15 of each 28-day cycle) plus oral vemurafenib (720 mg twice daily) plus oral cobimetinib (60 mg once daily, days 1-21); atezolizumab was withheld in cycle 1. Treatment was continued until progression, toxicity, or death. The primary outcome was intracranial objective response rate confirmed by assessments at least 4 weeks apart, as assessed by independent review committee (IRC) using modified Response Evaluation Criteria in Solid Tumours version 1.1. Because of early closure of the BRAFV600 wild-type cohort, the primary endpoint of intracranial objective response rate by IRC assessment was not done in this cohort; intracranial objective response rate by investigator assessment was reported instead. Efficacy and safety were analysed in all patients who received at least one dose of study medication. This trial is closed to enrolment and is registered with ClinicalTrials.gov, NCT03625141. FINDINGS Between Dec 13, 2018, and Dec 7, 2020, 65 patients were enrolled in the BRAFV600 mutation-positive cohort; the BRAFV600 wild-type cohort was closed early after enrolment of 15 patients. Median follow-up was 9·7 months (IQR 6·3-15·0) for the BRAFV600 mutation-positive cohort and 6·2 months (3·5-23·0) for the BRAFV600 wild-type cohort. Intracranial objective response rate was 42% (95% CI 29-54) by IRC assessment in the BRAFV600 mutation-positive cohort and 27% (95% CI 8-55) by investigator assessment in the BRAFV600 wild-type cohort. Treatment-related grade 3 or worse adverse events occurred in 41 (68%) of 60 patients who received atezolizumab plus vemurafenib plus cobimetinib in the BRAFV600 mutation-positive cohort, the most common of which were lipase increased (15 [25%] of 60 patients) and blood creatine phosphokinase increased (11 [18%]). Eight (53%) of 15 patients treated with atezolizumab plus cobimetinib in the BRAFV600 wild-type cohort had treatment-related grade 3 or worse adverse events, most commonly anaemia (two [13%]) and dermatitis acneiform (two [13%]). Treatment-related serious adverse events occurred in 14 (23%) of 60 patients who received triplet therapy in the BRAFV600 mutation-positive cohort and two (13%) of 15 in the BRAFV600 wild-type cohort. No treatment-related deaths occurred. INTERPRETATION Atezolizumab plus vemurafenib and cobimetinib provided intracranial activity in patients with BRAFV600-mutated melanoma with CNS metastases. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Reinhard Dummer
- Department of Dermatology, Skin Cancer Center, University Hospital Zurich, Zurich, Switzerland.
| | | | | | - Youyou Hu
- F Hoffman-La Roche, Basel, Switzerland
| | - Dao Wang
- F Hoffman-La Roche, Basel, Switzerland
| | - Sergio Jobim de Azevedo
- Hospital de Clínicas de Porto Alegre, Unidade de Pesquisa Clinica em Oncologia, Porto Alegre, Brazil
| | - Caroline Robert
- Gustave Roussy and Université Paris-Saclay, Villejuif-Paris, France
| | | | | | | | | | | | | | - Luis de la Cruz Merino
- Hospital Universitario Virgen Macarena, Clinical Oncology Department and Medicine Department, University of Seville, Seville, Spain
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
30
|
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: 207] [Impact Index Per Article: 103.5] [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
|
31
|
Goodman RS, Johnson DB, Balko JM. Corticosteroids and Cancer Immunotherapy. Clin Cancer Res 2023; 29:2580-2587. [PMID: 36648402 PMCID: PMC10349688 DOI: 10.1158/1078-0432.ccr-22-3181] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/22/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023]
Abstract
Despite revolutionizing cancer management, immunotherapies dysregulate the immune system, leading to immune-mediated adverse events. These common and potentially dangerous toxicities are often treated with corticosteroids, which are among the most prescribed drugs in oncology for a wide range of cancer and noncancer indications. While steroids exert several mechanisms to reduce immune activity, immunotherapies, such as immune checkpoint inhibitors (ICI), are designed to enhance the immune system's inherent antitumor activity. Because ICI requires an intact and robust immune response, the immunosuppressive properties of steroids have led to a widespread concern that they may interfere with antitumor responses. However, the existing data of the effect of systemic steroids on immunotherapy efficacy remain somewhat conflicted and unclear. To inform clinical decision-making and improve outcomes, we review the impact of steroids on antitumor immunity, recent advances in the knowledge of their impact on ICI efficacy in unique populations and settings, associated precautions, and steroid-sparing treatment approaches.
Collapse
Affiliation(s)
| | - Douglas B. Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Justin M. Balko
- Department of Medicine, Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
32
|
van Zeijl MCT, van Breeschoten J, de Wreede LC, Wouters MWJM, Hilarius DL, Blank CU, Aarts MJB, van den Berkmortel FWPJ, de Groot JWB, Hospers GAP, Kapiteijn E, Piersma D, van Rijn RS, Stevense-den Boer MA, van der Veldt AAM, Vreugdenhil G, Boers-Sonderen MJ, Suijkerbuijk KPM, Haanen JBAG, van den Eertwegh AJM. Real-world Outcomes of Ipilimumab Plus Nivolumab Combination Therapy in a Nation-wide Cohort of Advanced Melanoma Patients in the Netherlands. J Immunother 2023; 46:197-204. [PMID: 37103470 DOI: 10.1097/cji.0000000000000468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 04/28/2023]
Abstract
In phase III trials, ipilimumab plus nivolumab combination therapy is highly efficacious for advanced melanoma, despite many treatment-related grades 3-4 adverse events. Here, we report real-world safety and survival outcomes of ipilimumab plus nivolumab for advanced melanoma. Patients with advanced melanoma who received first-line ipilimumab plus nivolumab between January 1, 2015 and June 30, 2021 were selected from the Dutch Melanoma Treatment Registry. We evaluated response status at 3, 6, 12, 18, and 24 months. OS and PFS were estimated with the Kaplan-Meier method. Separate analyses were performed for patients with or without brain metastases and for patients who met the inclusion criteria of the Checkmate-067 trial. In total, 709 patients received first-line ipilimumab plus nivolumab. Three hundred sixty (50.7%) patients experienced grade 3-4 adverse events, with 211 of the (58.6%) patients requiring hospital admission. The median treatment duration was 42 days (IQR = 31-139). At 24 months, disease control was achieved in 37% of patients. Median PFS since the start of treatment was 6.6 months (95% CI: 5.3-8.7), and median OS was 28.7 months (95% CI: 20.7-42.2). CheckMate-067 trial-like patients had a 4-year OS of 50% (95% CI: 43-59). Among patients with no asymptomatic or symptomatic brain metastases, the 4-year OS probabilities were 48% (95% CI: 41-55), 45% (95% CI: 35-57), and 32% (95% CI: 23-46). Ipilimumab plus nivolumab can achieve long-term survival in advanced melanoma patients in a real-world setting, including patients not represented in the CheckMate-067 trial. However, the proportion of patients with disease control in the real world is lower compared with clinical trials.
Collapse
Affiliation(s)
- Michiel C T van Zeijl
- Scientific Department, Dutch Institute for Clinical Auditing
- Department of Medical Oncology, Leiden University Medical Centre
| | - Jesper van Breeschoten
- Scientific Department, Dutch Institute for Clinical Auditing
- Department of Medical Oncology, Amsterdam UMC-location VUmc, Cancer Center Amsterdam
| | - Liesbeth C de Wreede
- Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, The Netherlands
| | - Michel W J M Wouters
- Scientific Department, Dutch Institute for Clinical Auditing
- Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Surgical Oncology, Netherlands Cancer Institute
| | | | - Christian U Blank
- Divisions of Medical Oncology and Molecular Oncology & Immunology, Netherlands Cancer Institute
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam
| | - Maureen J B Aarts
- Department of Medical Oncology, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht
| | | | | | - Geke A P Hospers
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre
| | - Djura Piersma
- Department of Internal Medicine, Medisch Spectrum Twente, Enschede
| | | | | | - Astrid A M van der Veldt
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Centre, Rotterdam
| | | | | | | | - John B A G Haanen
- Department of Medical Oncology, Leiden University Medical Centre
- Divisions of Medical Oncology and Molecular Oncology & Immunology, Netherlands Cancer Institute
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam
| | | |
Collapse
|
33
|
Galldiks N, Lohmann P, Fink GR, Langen KJ. Amino Acid PET in Neurooncology. J Nucl Med 2023; 64:693-700. [PMID: 37055222 DOI: 10.2967/jnumed.122.264859] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/10/2023] [Indexed: 04/15/2023] Open
Abstract
For decades, several amino acid PET tracers have been used to optimize diagnostics in patients with brain tumors. In clinical routine, the most important clinical indications for amino acid PET in brain tumor patients are differentiation of neoplasm from nonneoplastic etiologies, delineation of tumor extent for further diagnostic and treatment planning (i.e., diagnostic biopsy, resection, or radiotherapy), differentiation of treatment-related changes such as pseudoprogression or radiation necrosis after radiation or chemoradiation from tumor progression at follow-up, and assessment of response to anticancer therapy, including prediction of patient outcome. This continuing education article addresses the diagnostic value of amino acid PET for patients with either glioblastoma or metastatic brain cancer.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany;
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany; and
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany; and
- Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
34
|
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
|
35
|
Nikoo M, Rabiee F, Mohebbi H, Eghbalifard N, Rajabi H, Yazdani Y, Sakhaei D, Khosravifarsani M, Akhavan-Sigari R. Nivolumab plus ipilimumab combination therapy in cancer: Current evidence to date. Int Immunopharmacol 2023; 117:109881. [PMID: 37012882 DOI: 10.1016/j.intimp.2023.109881] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 03/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer immunotherapy, yielding significant antitumor responses across multiple cancer types. Combination ICI therapy with anti-CTLA-4 and anti-PD-1 antibodies outperforms either antibody alone in terms of clinical efficacy. As a consequence, the U.S. Food and Drug Administration (FDA) approved ipilimumab (anti-CTLA-4) plus nivolumab (anti-PD-1) as the first-ever approved therapies for combined ICI in patients with metastatic melanoma. Despite the success of ICIs, treatment with checkpoint inhibitor combinations poses significant clinical challenges, such as increased rates of immune-related adverse events (irAEs) and drug resistance. Thus, identifying optimal prognostic biomarkers could help to monitor the safety and efficacy of ICIs and identify patients who may benefit the most from these treatments. In this review, we will first go over the fundamentals of the CTLA-4 and PD-1 pathways, as well as the mechanisms of ICI resistance. The results of clinical findings that evaluated the combination of ipilimumab and nivolumab are then summarized to support future research in the field of combination therapy. Finally, the irAEs associated with combined ICI therapy, as well as the underlying biomarkers involved in their management, are discussed.
Collapse
|
36
|
Straehla JP, Reardon DA, Wen PY, Agar NYR. The Blood-Brain Barrier: Implications for Experimental Cancer Therapeutics. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:265-289. [PMID: 38323268 PMCID: PMC10846865 DOI: 10.1146/annurev-cancerbio-061421-040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The blood-brain barrier is critically important for the treatment of both primary and metastatic cancers of the central nervous system (CNS). Clinical outcomes for patients with primary CNS tumors are poor and have not significantly improved in decades. As treatments for patients with extracranial solid tumors improve, the incidence of CNS metastases is on the rise due to suboptimal CNS exposure of otherwise systemically active agents. Despite state-of-the art surgical care and increasingly precise radiation therapy, clinical progress is limited by the ability to deliver an effective dose of a therapeutic agent to all cancerous cells. Given the tremendous heterogeneity of CNS cancers, both across cancer subtypes and within a single tumor, and the range of diverse therapies under investigation, a nuanced examination of CNS drug exposure is needed. With a shared goal, common vocabulary, and interdisciplinary collaboration, the field is poised for renewed progress in the treatment of CNS cancers.
Collapse
Affiliation(s)
- Joelle P Straehla
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Tringale KR, Reiner AS, Sehgal RR, Panageas K, Betof Warner AS, Postow MA, Moss NS. Efficacy of immunotherapy for melanoma brain metastases in patients with concurrent corticosteroid exposure. CNS Oncol 2023; 12:CNS93. [PMID: 36802833 PMCID: PMC9996406 DOI: 10.2217/cns-2022-0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Aim: Immune checkpoint inhibitor (ICI) efficacy is undefined for melanoma brain metastases (MBM) with concurrent corticosteroid exposure. Materials & methods: We retrospectively evaluated patients with untreated MBM who received corticosteroids (≥1.5 mg dexamethasone equivalent) within 30 days of ICI. mRECIST criteria and Kaplan-Meier methods defined intracranial progression-free survival (iPFS). The lesion size-response association was evaluated with repeated measures modeling. Results: A total of 109 MBM were evaluated. The patient level intracranial response rate was 41%. Median iPFS was 2.3 months and overall survival was 13.4 months. Larger lesions were more likely to progress, with diameter >2.05 cm most predictive of progression (OR: 18.9; 95% CI: 2.6-139.5; p = 0.004). There was no difference in iPFS with steroid exposure pre- versus post-ICI initiation. Conclusion: In the largest reported ICI+corticosteroid cohort, we identify size dependent MBM response.
Collapse
Affiliation(s)
- Kathryn R Tringale
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anne S Reiner
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ryka R Sehgal
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Katherine Panageas
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nelson S Moss
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Multidisciplinary Brain Metastasis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
38
|
Internò V, Sergi MC, Metta ME, Guida M, Trerotoli P, Strippoli S, Circelli S, Porta C, Tucci M. Melanoma Brain Metastases: A Retrospective Analysis of Prognostic Factors and Efficacy of Multimodal Therapies. Cancers (Basel) 2023; 15:1542. [PMID: 36900333 PMCID: PMC10001111 DOI: 10.3390/cancers15051542] [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: 12/19/2022] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Brain metastasis in cutaneous melanoma (CM) has historically been considered to be a dismal prognostic feature, although recent evidence has highlighted the intracranial activity of combined immunotherapy (IT). Herein, we completed a retrospective study to investigate the impact of clinical-pathological features and multimodal therapies on the overall survival (OS) of CM patients with brain metastases. A total of 105 patients were evaluated. Nearly half of the patients developed neurological symptoms leading to a negative prognosis (p = 0.0374). Both symptomatic and asymptomatic patients benefited from encephalic radiotherapy (eRT) (p = 0.0234 and p = 0.011). Lactate dehydrogenase (LDH) levels two times higher than the upper limit normal (ULN) at the time of brain metastasis onset was associated with poor prognosis (p = 0.0452) and identified those patients who did not benefit from eRT. Additionally, the poor prognostic role of LDH levels was confirmed in patients treated with targeted therapy (TT) (p = 0.0015) concerning those who received immunotherapy (IT) (p = 0.16). Based on these results, LDH levels higher than two times the ULN at the time of the encephalic progression identify those patients with a poor prognosis who did not benefit from eRT. The negative prognostic role of LDH levels on eRT observed in our study will require prospective evaluations.
Collapse
Affiliation(s)
- Valeria Internò
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70121 Bari, Italy
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria, Policlinico di Bari, 70124 Bari, Italy
| | - Maria Chiara Sergi
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria, Policlinico di Bari, 70124 Bari, Italy
| | - Maria Elvira Metta
- Medical Statistic and Biometry Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Michele Guida
- IRCCS, Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Paolo Trerotoli
- Medical Statistic and Biometry Unit, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | | | - Salvatore Circelli
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria, Policlinico di Bari, 70124 Bari, Italy
| | - Camillo Porta
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70121 Bari, Italy
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria, Policlinico di Bari, 70124 Bari, Italy
| | - Marco Tucci
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70121 Bari, Italy
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria, Policlinico di Bari, 70124 Bari, Italy
| |
Collapse
|
39
|
Vasudevan HN, Susko MS, Ma L, Nakamura JL, Raleigh DR, Boreta L, Fogh S, Theodosopoulos PV, McDermott MW, Tsai KK, Sneed PK, Braunstein SE. Mutational Status and Clinical Outcomes Following Systemic Therapy with or without Focal Radiation for Resected Melanoma Brain Metastases. World Neurosurg 2023; 170:e514-e519. [PMID: 36400359 DOI: 10.1016/j.wneu.2022.11.060] [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: 10/05/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Brain metastases occur frequently in advanced melanoma and traditionally require surgery and radiation therapy. New evidence demonstrates that systemic therapies are effective for controlling metastatic melanoma brain metastases. This study evaluated outcomes after resection of melanoma brain metastases treated with systemic therapy, with or without focal radiotherapy. METHODS All patients received immunotherapy or BRAF/MEK inhibitors preoperatively or in the immediate 3 months postoperatively. Resection cavity failure, distant central nervous system progression, and adverse radiation effects were reported in the presence and absence of focal radiotherapy using the Kaplan-Meier method. RESULTS Between 2011 and 2020, 37 resection cavities in 29 patients met criteria for analysis. Of lesions, 22 (59%) were treated with focal radiotherapy, and 15 (41%) were treated with targeted therapy or immunotherapy alone. The 12- and 24-month freedom from local recurrence was 64.8% (95% confidence interval [CI] 42.1%-99.8%) and 46.3% (95% CI 24.5%-87.5%), respectively, for systemic therapy alone and 93.3% (95% CI 81.5%-100%) at both time points for focal radiotherapy (P = 0.01). On univariate analysis, focal radiotherapy was the only significant factor associated with reduction of local recurrence risk (hazard ratio 0.10, 95% CI 0.01-0.85; P = 0.04). There were no significant differences in central nervous system progression-free survival or overall survival between patients who received systemic therapy plus focal radiotherapy compared with systemic therapy alone. BRAF mutation status was reviewed for either the brain metastasis (n = 9 patients, 31%) or the primary site (n = 20 patients, 69%), and patients harboring BRAFV600E mutations had worse progression-free survival (P = 0.043). CONCLUSIONS Focal radiotherapy with systemic therapy for resected melanoma brain metastases significantly decreased resection cavity recurrence compared with systemic therapy alone. BRAF mutation status correlated with poorer outcomes.
Collapse
Affiliation(s)
- Harish N Vasudevan
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Matthew S Susko
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Lijun Ma
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Jean L Nakamura
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA; Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Lauren Boreta
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Shannon Fogh
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Philip V Theodosopoulos
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Michael W McDermott
- Miami Neuroscience Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Katy K Tsai
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Penny K Sneed
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
40
|
Wilmott JS, Tawbi H, Engh JA, Amankulor N, Shivalingam B, Banerjee H, Vergara IA, Lee H, Johansson PA, Ferguson PM, Saiag P, Robert C, Grob JJ, Butterfield LH, Scolyer RA, Kirkwood JM, Long GV, Davies MA. Clinical Features Associated with Outcomes and Biomarker Analysis of Dabrafenib plus Trametinib Treatment in Patients with BRAF-Mutant Melanoma Brain Metastases. Clin Cancer Res 2023; 29:521-531. [PMID: 36477181 PMCID: PMC9898142 DOI: 10.1158/1078-0432.ccr-22-2581] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/04/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE This study aimed to identify baseline clinical features associated with the outcomes of patients enrolled in the COMBI-MB phase II study of dabrafenib and trametinib treatment in patients with V600 BRAF-mutant metastatic melanoma with melanoma brain metastases (MBM). Exploratory biomarker analysis was also conducted as part of the synergistic COMBI-BRV trial (BRV116521), to identify molecular and immunologic changes associated with dabrafenib in MBMs and extracranial metastases (ECM). PATIENTS AND METHODS Post hoc analysis was performed for baseline features of patients (n = 125) enrolled in COMBI-MB. Analyses were performed to identify baseline clinical features associated with intracranial response rate (ICRR), progression-free survival (PFS), and overall survival (OS). Exploratory biomarker analysis was performed on biospecimen collected in the COMBI-BRV trial in which patients with BRAF-mutant, resectable MBM were treated with dabrafenib for 10 to 14 days prior to craniotomy. Accessible ECM were resected or biopsied at the time of craniotomy. Biospecimens underwent molecular and immunologic profiling for comparative analyses. RESULTS In COMBI-MB baseline treatment with corticosteroids was independently associated with lower ICRR [39% vs. 63%; OR, 0.323; 95 % confidence interval (CI), 0.105-0.996; P = 0.049] and shorter PFS (HR, 1.93; 95% CI, 1.06-3.51; P = 0.031). Additional significant associations identified in the multivariate analysis were improved PFS in patients with a BRAFV600E genotype (HR, 0.565; 95% CI, 0.321-0.996; P = 0.048) and improved OS in patients with Eastern Cooperative Oncology Group 0 (HR, 0.44; 95% CI, 0.25-0.78; P = 0.005). CONCLUSIONS Corticosteroid treatment was associated with reduced ICRR and PFS in COMBI-MB, similar to results with immunotherapy for MBMs. Baseline corticosteroid treatment is a key factor to consider in MBM patient management and clinical trial design/interpretation.
Collapse
Affiliation(s)
- James S. Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Hussein Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Johnathan A Engh
- The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Nduka Amankulor
- The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Brindha Shivalingam
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia,Department of Neurosurgery, Royal Prince Alfred Hospital, NSW, Australia
| | - Hiya Banerjee
- Novartis Pharmaceuticals Corporation, Basel, Switzerland
| | - Ismael A. Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Hansol Lee
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Peter A. Johansson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Peter M Ferguson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Philippe Saiag
- Dermatology Department, Ambroise Paré Hospital, APHP, Versailles University – Paris-Saclay, Boulogne-Billancourt, France
| | - Caroline Robert
- Gustave Roussy and Paris Saclay University, Villejuif, France
| | | | - Lisa H. Butterfield
- The Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Richard A. Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - John M Kirkwood
- The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Georgina V. Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia,Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | |
Collapse
|
41
|
Castelo-Branco L, Morgan G, Prelaj A, Scheffler M, Canhão H, Van Meerbeeck JP, Awada A. Challenges and knowledge gaps with immune checkpoint inhibitors monotherapy in the management of patients with non-small-cell lung cancer: a survey of oncologist perceptions. ESMO Open 2023; 8:100764. [PMID: 36640544 PMCID: PMC10024152 DOI: 10.1016/j.esmoop.2022.100764] [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: 05/26/2022] [Revised: 11/13/2022] [Accepted: 12/05/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Immune checkpoint-inhibitors (ICIs) are changing outcomes in different cancer settings, notably for patients with non-small-cell lung cancer (NSCLC). There are, however, still important gaps of evidence for clinical practice when using these novel treatments. In this study, we assessed physicians' opinion and experience on challenges for clinical practice with ICIs monotherapy in NSCLC. METHODS A survey was conducted on experienced physicians treating patients with NSCLC with ICIs. Two rounds of pilot tests were carried out for validation among a group of experts. Topics under analysis were in relation to treatment of elderly populations, performance status, brain metastases, use of steroids or antibiotics, the effects of gut microbiome, autoimmune diseases, human immunodeficiency virus infection, solid organ transplants, use of anti-programmed cell death protein 1 versus anti-programmed death-ligand 1 drugs, atypical tumour responses, predictors of response, duration of treatment and a final open question on additional relevant challenges. RESULTS Two hundred and twenty-one answers were collected, including 106 (48%) valid answers from experts for final analysis (physicians who have treated at least 20 patients with NSCLC with ICIs). The vast majority agreed that the selected topics in this study are important challenges ahead and more evidence is needed. Moreover, predictors of response, treating brain metastasis, shorter duration of treatment, the effects of gut microbiome and concomitant use of steroids were voted the most important topics to be further addressed in prospective clinical research. CONCLUSIONS This survey contributed to understanding which are the main challenges for clinical practice with ICIs monotherapy in NSCLC. It can also contribute to guide further clinical research, considering the opinions and experience of those who regularly treat NSCLC patients with ICIs.
Collapse
Affiliation(s)
- L Castelo-Branco
- NOVA National School of Public Health, NOVA University, Lisbon, Portugal.
| | - G Morgan
- Skåne University Hospital, Division of Medical and Radiation Oncology, Lund, Sweden
| | - A Prelaj
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - M Scheffler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Cologne, Germany
| | - H Canhão
- EPIDOC Unit, Comprehensive Health Research Center (CHRC), NOVA Medical School, NOVA University, Lisbon; Centro Hospitalar Universitario Lisboa Central, Lisbon, Portugal
| | | | - A Awada
- Oncology Medicine Department, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
42
|
Patient-specific identification of genome-wide DNA-methylation differences between intracranial and extracranial melanoma metastases. Sci Rep 2023; 13:444. [PMID: 36624125 PMCID: PMC9829750 DOI: 10.1038/s41598-022-24940-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/22/2022] [Indexed: 01/11/2023] Open
Abstract
Melanomas frequently metastasize to distant organs and especially intracranial metastases still represent a major clinical challenge. Epigenetic reprogramming of intracranial metastases is thought to be involved in therapy failure, but so far only little is known about patient-specific DNA-methylation differences between intra- and extracranial melanoma metastases. Hierarchical clustering of the methylomes of 24 patient-matched intra- and extracranial melanoma metastases pairs revealed that intra- and extracranial metastases of individual patients were more similar to each other than to metastases in the same tissue from other patients. Therefore, a personalized analysis of each metastases pair was done by a Hidden Markov Model to classify methylation levels of individual CpGs as decreased, unchanged or increased in the intra- compared to the extracranial metastasis. The predicted DNA-methylation alterations were highly patient-specific differing in the number and methylation states of altered CpGs. Nevertheless, four important general observations were made: (i) intracranial metastases of most patients mainly showed a reduction of DNA-methylation, (ii) cytokine signaling was most frequently affected by differential methylation in individual metastases pairs, but also MAPK, PI3K/Akt and ECM signaling were often altered, (iii) frequently affected genes were mainly involved in signaling, growth, adhesion or apoptosis, and (iv) an enrichment of functional terms related to channel and transporter activities supports previous findings for a brain-like phenotype. In addition, the derived set of 17 signaling pathway genes that distinguished intra- from extracranial metastases in more than 50% of patients included well-known oncogenes (e.g. PRKCA, DUSP6, BMP4) and several other genes known from neuronal disorders (e.g. EIF4B, SGK1, CACNG8). Moreover, associations of gene body methylation alterations with corresponding gene expression changes revealed that especially the three signaling pathway genes JAK3, MECOM, and TNXB differ strongly in their expression between patient-matched intra- and extracranial metastases. Our analysis contributes to an in-depth characterization of DNA-methylation differences between patient-matched intra- and extracranial melanoma metastases and may provide a basis for future experimental studies to identify targets for new therapeutic approaches.
Collapse
|
43
|
Switzer B, Piperno-Neumann S, Lyon J, Buchbinder E, Puzanov I. Evolving Management of Stage IV Melanoma. Am Soc Clin Oncol Educ Book 2023; 43:e397478. [PMID: 37141553 DOI: 10.1200/edbk_397478] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Significant advancements have been made in the treatment of advanced melanoma with the use of immune checkpoint inhibitors, novel immunotherapies, and BRAF/MEK-targeted therapies with numerous frontline treatment options. However, there remains suboptimal evidence to guide treatment decisions in many patients. These include patients with newly diagnosed disease, immune checkpoint inhibitor (ICI)-resistant/ICI-refractory disease, CNS metastases, history of autoimmune disease, and/or immune-related adverse events (irAEs). Uveal melanoma (UM) is a rare melanoma associated with a poor prognosis in the metastatic setting. Systemic treatments, including checkpoint inhibitors, failed to demonstrate any survival benefit. Tebentafusp, a bispecific molecule, is the first treatment to improve overall survival (OS) in patients with HLA A*02:01-positive metastatic UM.
Collapse
Affiliation(s)
- Benjamin Switzer
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | - James Lyon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| |
Collapse
|
44
|
Sharma P, Aaroe A, Liang J, Puduvalli VK. Tumor microenvironment in glioblastoma: Current and emerging concepts. Neurooncol Adv 2023; 5:vdad009. [PMID: 36968288 PMCID: PMC10034917 DOI: 10.1093/noajnl/vdad009] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Glioblastoma (GBM) tumor microenvironment (TME) is a highly heterogeneous and complex system, which in addition to cancer cells, consists of various resident brain and immune cells as well as cells in transit through the tumor such as marrow-derived immune cells. The TME is a dynamic environment which is heavily influenced by alterations in cellular composition, cell-to-cell contact and cellular metabolic products as well as other chemical factors, such as pH and oxygen levels. Emerging evidence suggests that GBM cells appear to reprogram their the TME, and hijack microenvironmental elements to facilitate rapid proliferation, invasion, migration, and survival thus generating treatment resistance. GBM cells interact with their microenvironment directly through cell-to-cell by interaction mediated by cell-surface molecules, or indirectly through apocrine or paracrine signaling via cytokines, growth factors, and extracellular vehicles. The recent discovery of neuron-glioma interfaces and neurotransmitter-based interactions has uncovered novel mechanisms that favor tumor cell survival and growth. Here, we review the known and emerging evidence related to the communication between GBM cells and various components of its TME, discuss models for studying the TME and outline current studies targeting components of the TME for therapeutic purposes.
Collapse
Affiliation(s)
- Pratibha Sharma
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ashley Aaroe
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jiyong Liang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vinay K Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
45
|
Tabor JK, Onoichenco A, Narayan V, Wernicke AG, D’Amico RS, Vojnic M. Brain metastasis screening in the molecular age. Neurooncol Adv 2023; 5:vdad080. [PMID: 37484759 PMCID: PMC10358433 DOI: 10.1093/noajnl/vdad080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
The incidence of brain metastases (BM) amongst cancer patients has been increasing due to improvements in therapeutic options and an increase in overall survival. Molecular characterization of tumors has provided insights into the biology and oncogenic drivers of BM and molecular subtype-based screening. Though there are currently some screening and surveillance guidelines for BM, they remain limited. In this comprehensive review, we review and present epidemiological data on BM, their molecular characterization, and current screening guidelines. The molecular subtypes with the highest BM incidence are epithelial growth factor receptor-mutated non-small cell lung cancer (NSCLC), BRCA1, triple-negative (TN), and HER2+ breast cancers, and BRAF-mutated melanoma. Furthermore, BMs are more likely to present asymptomatically at diagnosis in oncogene-addicted NSCLC and BRAF-mutated melanoma. European screening standards recommend more frequent screening for oncogene-addicted NSCLC patients, and clinical trials are investigating screening for BM in hormone receptor+, HER2+, and TN breast cancers. However, more work is needed to determine optimal screening guidelines for other primary cancer molecular subtypes. With the advent of personalized medicine, molecular characterization of tumors has revolutionized the landscape of cancer treatment and prognostication. Incorporating molecular characterization into BM screening guidelines may allow physicians to better identify patients at high risk for BM development and improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Vinayak Narayan
- Department of Neurological Surgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - A Gabriella Wernicke
- Department of Radiation Medicine, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - Randy S D’Amico
- Department of Neurological Surgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - Morana Vojnic
- Corresponding Author: Morana Vojnic, MD, MBA, 210 East 64th Street, Floor 4, New York, NY 10065, USA ()
| |
Collapse
|
46
|
Shui IM, Scherrer E, Frederickson A, Li JW, Mynzhassarova A, Druyts E, Tawbi H. Resistance to anti-PD1 therapies in patients with advanced melanoma: systematic literature review and application of the Society for Immunotherapy of Cancer Immunotherapy Resistance Taskforce anti-PD1 resistance definitions. Melanoma Res 2022; 32:393-404. [PMID: 36223314 DOI: 10.1097/cmr.0000000000000850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nearly half of advanced melanoma patients do not achieve a clinical response with anti-programmed cell death 1 protein (PD1) therapy (i.e. primary resistance) or initially achieve a clinical response but eventually progress during or following further treatment (i.e. secondary resistance). A consensus definition for tumor resistance to anti-PD1 monotherapy was published by Society for Immunotherapy of Cancer Immunotherapy Resistance Taskforce (SITC) in 2020. A systematic literature review (SLR) of clinical trials and observational studies was conducted to characterize the proportions of advanced melanoma patients who have progressed on anti-PD1 therapies. The SLR included 55 unique studies and the SITC definition of primary resistance was applied to 37 studies that specified disease progression by best overall response. Median and range of patients with primary resistance in studies that specified first-line and second-line or higher anti-PD1 monotherapy was 35.50% (21.19-39.13%; n = 4 studies) and 41.54% (30.00-56.41%, n = 3 studies); median and range of patients with primary resistance in studies that specified first-line and second-line or higher combination therapy was 30.23% (15.79-33.33%; n = 6 studies), and 70.00% (61.10-73.33%; n = 3 studies). Primary resistance to anti-PD1 monotherapies and when in combination with ipilimumab are higher in patients receiving second-line or higher therapies, in patients with acral, mucosal, and uveal melanoma, and in patients with active brain metastases. The percentage of patients with primary resistance was generally consistent across clinical trials, with variability in resistance noted for observational studies. Limitations include applying the SITC definitions to combination therapies, where consensus definitions are not yet available. Future studies should highly consider utilizing the SITC definitions to harmonize how resistance is classified and facilitate meaningful context for clinical activity.
Collapse
Affiliation(s)
| | | | | | - Joyce W Li
- Pharmalytics Group, Vancouver, British Columbia, Canada
| | | | - Eric Druyts
- Pharmalytics Group, Vancouver, British Columbia, Canada
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
47
|
The Analysis of Trends in Survival for Patients with Melanoma Brain Metastases with Introduction of Novel Therapeutic Options before the Era of Combined Immunotherapy-Multicenter Italian-Polish Report. Cancers (Basel) 2022; 14:cancers14235763. [PMID: 36497248 PMCID: PMC9737166 DOI: 10.3390/cancers14235763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Stage IV melanoma patients develop melanoma brain metastases (MBM) in 50% of cases. Their prognosis is improving, and its understanding outside the context of clinical trials is relevant. We have retrospectively analyzed the clinical data, course of treatment, and outcomes of 531 subsequent stage IV melanoma patients with BM treated in five reference Italian and Polish melanoma centers between 2014 and 2021. Patients with MBM after 2017 had a better prognosis, with a significantly improved median of overall survival (OS) after 2017 in the worst mol-GPA prognostic groups (mol-GPA ≤ 2): a median OS >6 months and HR 0.76 vs. those treated before 2017 (CI: 0.60−0.97, p = 0.027). In our prognostic model, mol-GPA was highly predictive for survival, and symptoms without steroid use did not have prognostic significance. Local therapy significantly improved survival regardless of the year of diagnosis (treated before or after 2017), with median survival >12 months. Systemic therapy improved outcomes when it was combined with local therapy. Local surgery was associated with improved OS regardless of the timing related to treatment start (i.e., before or after 30 days from MBM diagnosis). Local and systemic treatment significantly prolong survival for the poorest mol-GPA prognosis. Use of modern treatment modalities is justified in all mol-GPA prognostic groups.
Collapse
|
48
|
Dummer R, Tawbi H. Symptomatic melanoma CNS metastases in the TRICOTEL study – Authors' reply. Lancet Oncol 2022; 23:e482. [DOI: 10.1016/s1470-2045(22)00647-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/30/2022]
|
49
|
Serra-Bellver P, Versluis JM, Oberoi HK, Zhou C, Slattery TD, Khan Y, Patrinely JR, Pires da Silva I, Martínez-Vila C, Cook N, Graham DM, Carlino MS, Menzies AM, Arance AM, Johnson DB, Long GV, Pickering L, Larkin JMG, Blank CU, Lorigan P. Real-world outcomes with ipilimumab and nivolumab in advanced melanoma: a multicentre retrospective study. Eur J Cancer 2022; 176:121-132. [PMID: 36215945 DOI: 10.1016/j.ejca.2022.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE To assess efficacy and toxicity of combination immunotherapy with ipilimumab plus nivolumab in routine practice in a retrospective multicentre cohort of patients with advanced melanoma. PATIENTS AND METHODS This retrospective analysis included patients with advanced melanoma treated with ipilimumab and nivolumab between October 2015 and January 2020 at six centres in Australia, Europe and the United States of America. We describe efficacy outcomes (overall survival [OS], progression-free survival [PFS] and objective response rate [ORR]) in treatment-naïve and pre-treated patients, with and without brain metastases, plus treatment-related adverse events (trAEs) in all patients treated. RESULTS A total of 697 patients were identified; 472 were treatment-naïve of which 138 (29.2%) had brain metastases, and 225 were previously treated of which 102 (45.3%) had brain metastases. At baseline, 32.3% had stage M1c and 34.4% stage M1d disease. Lactate dehydrogenase was high in 280 patients (40.2%). With a median follow-up of 25.9 months, median OS in the 334 treatment-naïve patients without brain metastases was 53.7 months (95% confidence interval [CI] 40.8-NR) and 38.7 months (95% CI 18.6-NR) for the 138 treatment-naïve patients with brain metastases. For the entire cohort the ORR was 48%, for treatment-naïve patients without brain metastases ORR was 56.6% with a median PFS of was 13.7 months (95% CI 9.6-26.5). Median PFS was 7.9 months (95% CI 5.8-10.4) and OS 38 months (95% CI 31-NR) for the entire cohort. Grade 3-4 trAE were reported in 44% of patients, and 4 (0.7%) treatment-related deaths (1 pneumonitis, 2 myocarditis and 1 colitis) were recorded. CONCLUSION The outcome and toxicity of combination immunotherapy with ipilimumab and nivolumab in a real-world patient population are similar to those reported in pivotal trials.
Collapse
Affiliation(s)
- Patricio Serra-Bellver
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom.
| | - Judith M Versluis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Honey K Oberoi
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Cong Zhou
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, United Kingdom
| | - Timothy D Slattery
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Yasir Khan
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - James R Patrinely
- Department of Medical Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Inês Pires da Silva
- Melanoma Institute Australia, The University of Sydney and the Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Westmead and Blacktown Hospital, Sydney, Australia
| | - C Martínez-Vila
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Natalie Cook
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, United Kingdom
| | - Donna M Graham
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, United Kingdom
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney and the Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Westmead and Blacktown Hospital, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney and the Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Ana M Arance
- Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Douglas B Johnson
- Department of Medical Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney and the Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Lisa Pickering
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - James M G Larkin
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paul Lorigan
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, United Kingdom
| |
Collapse
|
50
|
Eroglu Z, Topcu T, Yu H, Margolin K. How I treat brain metastases of melanoma. ESMO Open 2022; 7:100598. [PMID: 36274439 PMCID: PMC9589018 DOI: 10.1016/j.esmoop.2022.100598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 12/30/2022] Open
Abstract
Brain metastases are common in advanced melanoma and cause death in >50% of patients. Until recently, median survival was only ∼4 months. Improved systemic treatment including immune checkpoint inhibitors and combinations of BRAF/MEK inhibitors, however, has significantly improved intracranial tumor response and survival. In addition, advances in radiation therapy have also improved the intracranial outcomes for advanced melanoma patients with brain metastases (MBM). There has long been concern that systemic treatment of the central nervous metastases would be ineffective due to inability of active agents to cross an intact blood-brain barrier. Recent studies have shown, however, that highly active systemic therapy can have significant benefit in these patients. When determining a patient's treatment, the important factors in predicting the likelihood of benefit including the presence of neurologic symptoms, the number and size of brain metastases, performance status/status of extracranial disease, and BRAF mutation status should all be considered. In this review, we will discuss the challenges and treatment options for patients with advanced melanoma and brain metastases.
Collapse
Affiliation(s)
- Z. Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, USA,Correspondence to: Prof. Zeynep Eroglu, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612, USA. Tel: +1-813-745-4673
| | - T.O. Topcu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, USA
| | - H.M. Yu
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, USA
| | - K.A. Margolin
- Department of Medical Oncology, Providence St. John’s Cancer Institute, Santa Monica, USA
| |
Collapse
|