1
|
Elmusrati A, Wang CY. The expression of immune checkpoint proteins PD-L1 and TIM3 in mouse and human head and neck squamous cell carcinoma. Eur J Oral Sci 2024; 132:e13010. [PMID: 39090710 PMCID: PMC11436301 DOI: 10.1111/eos.13010] [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: 05/06/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
The aim of this study was to examine the expression of programmed death-ligand 1 (PD-L1) and of T cell immunoglobulin and mucin domain-containing protein (TIM3) in oral epithelial dysplasia and head and neck squamous cell carcinoma (HNSCC). Mouse HNSCC was induced with 4-nitroquinoline-1 oxide (4NQO). Oral epithelial dysplastic lesions, carcinoma in situ and HNSCC lesions were stained with anti-PD-L1 and TIM3 antibodies. The expression of PD-L1 and TIM3 in tumor cells and immune cells was semiquantitatively measured and compared. In parallel, human dysplasia and HNSCC were stained with anti-PD-L1 and anti-TIM3. The expression pattern of PD-L1+ and TIM3+ cells was further compared. In human and mouse samples both PD-L1 and TIM3 were found to be expressed in neoplastic and immune cells in HNSCC, but not in dysplasia. There was no significant difference in PD-L1 and TIM3 expression between metastatic and nonmetastatic HNSCC. We conclude that the 4NQO-induced mouse HNSCC model may be an excellent preclinical model for immune checkpoint therapy.
Collapse
Affiliation(s)
- Areeg Elmusrati
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Cun-Yu Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Lentz RW, Friedrich TJ, Blatchford PJ, Jordan KR, Pitts TM, Robinson HR, Davis SL, Kim SS, Leal AD, Lee MR, Waring MR, Martin AC, Dominguez AT, Bagby SM, Hartman SJ, Eckhardt SG, Messersmith WA, Lieu CH. A Phase II Study of Potentiation of Pembrolizumab with Binimetinib and Bevacizumab in Refractory Microsatellite-Stable Colorectal Cancer. Clin Cancer Res 2024; 30:3768-3778. [PMID: 38869830 PMCID: PMC11369619 DOI: 10.1158/1078-0432.ccr-24-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE In this single-institution phase II investigator-initiated study, we assessed the ability of MAPK and VEGF pathway blockade to overcome resistance to immunotherapy in microsatellite-stable metastatic colorectal cancer (MSS mCRC). PATIENTS AND METHODS Patients with MSS, BRAF wild-type mCRC who progressed on ≥2 prior lines of therapy received pembrolizumab, binimetinib, and bevacizumab until disease progression or unacceptable toxicity. After a safety run-in, patients were randomized to a 7-day run-in of binimetinib or simultaneous initiation of all study drugs, to explore whether MEK inhibition may increase tumor immunogenicity. The primary endpoint was objective response rate (ORR) in all patients combined (by Response Evaluation Criteria in Solid Tumors v1.1). RESULTS Fifty patients received study drug treatment; 54% were male with a median age of 55 years (range, 31-79). The primary endpoint, ORR, was 12.0% [95% confidence interval (CI) 4.5%-24.3%], which was not statistically different than the historical control data of 5% (P = 0.038, exceeding prespecified threshold of 0.025). The disease control rate was 70.0% (95% CI, 55.4%-82.1%), the median progression-free survival 5.9 months (95% CI, 4.2-8.7 months), and the median overall survival 9.3 months (95% CI, 6.7-12.2 months). No difference in efficacy was observed between the randomized cohorts. Grade 3 and 4 adverse events were observed in 56% and 8% of patients, respectively; the most common were rash (12%) and increased aspartate aminotransferase (12%). CONCLUSIONS Pembrolizumab, binimetinib, and bevacizumab failed to meet its primary endpoint of higher ORR compared with historical control data, demonstrated a high disease control rate, and demonstrated acceptable tolerability in refractory MSS mCRC.
Collapse
Affiliation(s)
- Robert W. Lentz
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Tyler J. Friedrich
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Patrick J. Blatchford
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Kimberly R. Jordan
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado.
| | - Todd M. Pitts
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Hannah R. Robinson
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - S. Lindsey Davis
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Sunnie S. Kim
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Alexis D. Leal
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Mathew R. Lee
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Meredith R.N. Waring
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Anne C. Martin
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Adrian T.A. Dominguez
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Stacey M. Bagby
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Sarah J. Hartman
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - S. Gail Eckhardt
- Department of Oncology, The University of Texas at Austin Dell Medical School, Austin, Texas.
| | - Wells A. Messersmith
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Christopher H. Lieu
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
3
|
Long GV, Carlino MS, Au-Yeung G, Spillane AJ, Shannon KF, Gyorki DE, Hsiao E, Kapoor R, Thompson JR, Batula I, Howle J, Ch'ng S, Gonzalez M, Saw RPM, Pennington TE, Lo SN, Scolyer RA, Menzies AM. Neoadjuvant pembrolizumab, dabrafenib and trametinib in BRAF V600-mutant resectable melanoma: the randomized phase 2 NeoTrio trial. Nat Med 2024; 30:2540-2548. [PMID: 38907159 PMCID: PMC11405264 DOI: 10.1038/s41591-024-03077-5] [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: 03/04/2024] [Accepted: 05/17/2024] [Indexed: 06/23/2024]
Abstract
Immune checkpoint inhibitors and BRAF-targeted therapy each improve survival in melanoma. Immune changes early during targeted therapy suggest the mechanisms of each drug class could work synergistically. In the non-comparative, randomized, phase 2 NeoTrio trial, we investigated whether targeted therapy could boost the proportion of patients achieving long-term recurrence-free survival with neoadjuvant immunotherapy in resectable stage III BRAFV600-mutant melanoma. Sixty patients (42% females) were randomized to pembrolizumab alone (n = 20), sequential therapy (dabrafenib plus trametinib followed by pembrolizumab; n = 20) or concurrent (triple) therapy (n = 20), followed by surgery and adjuvant therapy. The primary outcome was pathological response; secondary outcomes included radiographic response, recurrence-free survival, overall survival, surgical outcomes, peripheral blood and tumor analyses and safety. The pathological response rate was 55% (11/20; including six pathological complete responses (pCRs)) with pembrolizumab, 50% (10/20; three pCRs) with sequential therapy and 80% (16/20; ten pCRs) with concurrent therapy, which met the primary outcome in each arm. Treatment-related adverse events affected 75-100% of patients during neoadjuvant treatment, with seven early discontinuations (all in the concurrent arm). At 2 years, event-free survival was 60% with pembrolizumab, 80% with sequential therapy and 71% with concurrent therapy. Recurrences after major pathological response were more common in the targeted therapy arms, suggesting a reduction in response 'quality' when targeted therapy is added to neoadjuvant immunotherapy. Risking the curative potential of immunotherapy in melanoma cannot be justified. Pending longer follow-up, we suggest that immunotherapy and targeted therapy should not be combined in the neoadjuvant setting for melanoma. ClinicalTrials.gov registration: NCT02858921 .
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
- Royal North Shore Hospital, Sydney, New South Wales, Australia.
- Mater Hospital, Sydney, New South Wales, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Westmead Hospital, Westmead, New South Wales, Australia
- Blacktown Hospital, Blacktown, New South Wales, Australia
| | - George Au-Yeung
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew J Spillane
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
| | - Kerwin F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
- Concord Repatriation Hospital, Concord, New South Wales, Australia
| | - David E Gyorki
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Edward Hsiao
- Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Rony Kapoor
- Mater Hospital, Sydney, New South Wales, Australia
- I-MED Radiology Network, Mater Hospital, Sydney, New South Wales, Australia
| | - Jake R Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Iris Batula
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Julie Howle
- Westmead Hospital, Westmead, New South Wales, Australia
| | - Sydney Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Maria Gonzalez
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Thomas E Pennington
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Menzies AM, Lo SN, Saw RPM, Gonzalez M, Ch'ng S, Nieweg OE, Shannon KF, Ferguson PM, Lee J, Emmett L, Kapoor R, Rawson RV, Stretch JR, Thompson JF, Spillane AJ, Rizos H, Scolyer RA, Long GV. Five-year analysis of neoadjuvant dabrafenib and trametinib for stage III melanoma. Ann Oncol 2024; 35:739-746. [PMID: 38754780 DOI: 10.1016/j.annonc.2024.05.002] [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: 03/11/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Neoadjuvant dabrafenib plus trametinib has a high pathological response rate and impressive short-term survival in patients with resectable stage III melanoma. We report 5-year outcomes from the phase II NeoCombi trial. PATIENTS AND METHODS NeoCombi (NCT01972347) was a single-arm, open-label, single-centre, phase II trial. Eligible patients were adults (aged ≥18 years) with histologically confirmed, resectable, RECIST-measurable, American Joint Committee on Cancer seventh edition clinical stage IIIB-C BRAF V600E/K-mutant melanoma and Eastern Cooperative Oncology Group performance status ≤1. Patients received 52 weeks of treatment with dabrafenib 150 mg (orally twice per day) plus trametinib 2 mg (orally once per day), with complete resection of the pre-therapy tumour bed at week 12. RESULTS Between 20 August 2014 and 19 April 2017, 35 patients were enrolled. At data cut-off (17 August 2021), the median follow-up was 60 months [95% confidence interval (CI) 56-72 months]. Overall, 21 of 35 (60%) patients recurred, including 12 (57%) with first recurrence in locoregional sites (followed by later distant recurrence in 6) and 9 (43%) with first recurrence in distant sites, including 3 in the brain. Most recurrences occurred within 2 years, with no recurrences beyond 3 years. At 5 years, recurrence-free survival (RFS) was 40% (95% CI 27% to 60%), distant metastasis-free survival (DMFS) was 57% (95% CI 42% to 76%), and overall survival was 80% (95% CI 67% to 94%). Five-year survival outcomes were stratified by pathological response: RFS was 53% with pathological complete response (pCR) versus 28% with non-pCR (P = 0.087), DMFS was 59% versus 55% (P = 0.647), and overall survival was 88% versus 71% (P = 0.205), respectively. CONCLUSIONS Neoadjuvant dabrafenib plus trametinib has high pathological response rates in clinical stage III melanoma, but low rates of RFS, similar to those achieved with adjuvant targeted therapy alone. Patients with a pCR to dabrafenib plus trametinib still had a high risk of recurrence, unlike that seen with immunotherapy where recurrences are rare.
Collapse
Affiliation(s)
- A M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Royal North Shore Hospital, Sydney; Mater Hospital, Sydney.
| | - S N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney
| | - R P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Mater Hospital, Sydney; Royal Prince Alfred Hospital, Sydney
| | - M Gonzalez
- Melanoma Institute Australia, The University of Sydney, Sydney
| | - S Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Mater Hospital, Sydney; Royal Prince Alfred Hospital, Sydney
| | - O E Nieweg
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Mater Hospital, Sydney; Royal Prince Alfred Hospital, Sydney
| | - K F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Mater Hospital, Sydney; Royal Prince Alfred Hospital, Sydney
| | - P M Ferguson
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Royal Prince Alfred Hospital, Sydney
| | - J Lee
- Royal Prince Alfred Hospital, Sydney; Macquarie University, Sydney
| | | | | | - R V Rawson
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Royal Prince Alfred Hospital, Sydney
| | - J R Stretch
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Mater Hospital, Sydney; Royal Prince Alfred Hospital, Sydney
| | - J F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Mater Hospital, Sydney; Royal Prince Alfred Hospital, Sydney
| | - A J Spillane
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Royal North Shore Hospital, Sydney; Mater Hospital, Sydney
| | - H Rizos
- Melanoma Institute Australia, The University of Sydney, Sydney; Macquarie University, Sydney
| | - R A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Royal Prince Alfred Hospital, Sydney; Charles Perkins Centre, The University of Sydney, Sydney; NSW Health Pathology, Sydney, Australia
| | - G V Long
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Royal North Shore Hospital, Sydney; Mater Hospital, Sydney; Charles Perkins Centre, The University of Sydney, Sydney
| |
Collapse
|
5
|
Zhou L, Wu J, Ruan M, Xiao Y, Lan H, Wu Q, Yu CW, Zhang Q. The loss of B7-H4 expression in breast cancer cells escaping from T cell cytotoxicity contributes to epithelial-to-mesenchymal transition. Breast Cancer Res 2023; 25:115. [PMID: 37794509 PMCID: PMC10548745 DOI: 10.1186/s13058-023-01721-5] [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: 02/21/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND B7 homology 4 (B7-H4), a potential target for cancer therapy, has been demonstrated to inhibit T cell cytotoxicity in the early stages of breast cancer. However, B7-H4 manipulating breast tumor immune microenvironment (TIME) in the tumor progression remains unknown. METHODS We engineered T cells with B7-H4-specific chimeric antigen receptors (CARs) and performed a T cell co-culture assay to characterize B7-H4 expression level in breast cancer cells escaping from T cell cytotoxicity. We generated B7-H4 knockout (KO) and overexpression (OE) breast cancer cells to determine the epithelial-to-mesenchymal transition (EMT) and stemness characteristics in vitro and in vivo, including tumor proliferation, migration, metastasis and chemoresistance. The Cancer Genome Atlas breast cancer database was accessed to investigate the correlation between B7-H4 expression levels and EMT characteristics in patients with breast cancer. RESULTS Our result found that B7-H4 expression level was significantly reduced in a subset of breast cancer cells that escaped from the cytotoxicity of B7-H4 CAR-T cells. Compared with wild type cells, B7-H4 KO cells prompt EMT and stemness characteristics, including migration, invasion and metastasis, and OE cells vice versa. The increase in H3K27me3 in KO cells confirmed the epigenetic reprogramming of cancer stem cells. The IC50 of doxorubicin or oxaliplatin significantly increased in KO cells, which was in agreement with a decrease in OE cells. Moreover, a trend of downregulated B7-H4 from stage I to stage II breast cancer patients indicates that the low-expressing B7-H4 breast cancer cells escaping from TIME have spread to nearby breast lymph nodes in the cancer progression. CONCLUSIONS Our study illuminates the novel role of renouncing B7-H4 in breast cancer cells through immune escape, which contributes to EMT processes and provides new insights for breast cancer treatments.
Collapse
Affiliation(s)
- Linlin Zhou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jichun Wu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Mei Ruan
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Yonglei Xiao
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Hailin Lan
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Qiongwen Wu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Chen-Wei Yu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
- Department of Statistics and Information Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Qiuyu Zhang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
6
|
Castellani G, Buccarelli M, Arasi MB, Rossi S, Pisanu ME, Bellenghi M, Lintas C, Tabolacci C. BRAF Mutations in Melanoma: Biological Aspects, Therapeutic Implications, and Circulating Biomarkers. Cancers (Basel) 2023; 15:4026. [PMID: 37627054 PMCID: PMC10452867 DOI: 10.3390/cancers15164026] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer resulting from the malignant transformation of melanocytes. Recent therapeutic approaches, including targeted therapy and immunotherapy, have improved the prognosis and outcome of melanoma patients. BRAF is one of the most frequently mutated oncogenes recognised in melanoma. The most frequent oncogenic BRAF mutations consist of a single point mutation at codon 600 (mostly V600E) that leads to constitutive activation of the BRAF/MEK/ERK (MAPK) signalling pathway. Therefore, mutated BRAF has become a useful target for molecular therapy and the use of BRAF kinase inhibitors has shown promising results. However, several resistance mechanisms invariably develop leading to therapeutic failure. The aim of this manuscript is to review the role of BRAF mutational status in the pathogenesis of melanoma and its impact on differentiation and inflammation. Moreover, this review focuses on the mechanisms responsible for resistance to targeted therapies in BRAF-mutated melanoma and provides an overview of circulating biomarkers including circulating tumour cells, circulating tumour DNA, and non-coding RNAs.
Collapse
Affiliation(s)
- Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Beatrice Arasi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Elena Pisanu
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Bellenghi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| |
Collapse
|
7
|
Thavaneswaran S, Kansara M, Lin F, Espinoza D, Grady JP, Lee CK, Ballinger ML, Sebastian L, Corpuz T, Qiu MR, Mundra P, Bailey CG, Schmitz U, Simes J, Joshua AM, Thomas DM. A signal-seeking Phase 2 study of olaparib and durvalumab in advanced solid cancers with homologous recombination repair gene alterations. Br J Cancer 2023; 129:475-485. [PMID: 37365284 PMCID: PMC10403555 DOI: 10.1038/s41416-023-02311-0] [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: 10/20/2022] [Revised: 05/08/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
PURPOSE To determine the safety and efficacy of PARP plus PD-L1 inhibition (olaparib + durvalumab, O + D) in patients with advanced solid, predominantly rare cancers harbouring homologous recombination repair (HRR) defects. PATIENTS AND METHODS In total, 48 patients were treated with O + D, 16 with BRCA1/2 alterations (group 1) and 32 with other select HRR alterations (group 2). Overall, 32 (66%) patients had rare or less common cancers. The primary objective of this single-arm Phase II trial was a progression-free survival rate at 6 months (PFS6). Post hoc exploratory analyses were conducted on archival tumour tissue and serial bloods. RESULTS The PFS6 rate was 35% and 38% with durable objective tumour responses (OTR) in 3(19%) and 3(9%) in groups 1 and 2, respectively. Rare cancers achieving an OTR included cholangiocarcinoma, perivascular epithelioid cell (PEComa), neuroendocrine, gallbladder and endometrial cancer. O + D was safe, with five serious adverse events related to the study drug(s) in 3 (6%) patients. A higher proportion of CD38 high B cells in the blood and higher CD40 expression in tumour was prognostic of survival. CONCLUSIONS O + D demonstrated no new toxicity concerns and yielded a clinically meaningful PFS6 rate and durable OTRs across several cancers with HRR defects, including rare cancers.
Collapse
Affiliation(s)
- Subotheni Thavaneswaran
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia.
- The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia.
- Garvan Institute of Medical Research, Sydney, NSW, Australia.
| | - Maya Kansara
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Frank Lin
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Kinghorn Centre for Clinical Genomics, Sydney, NSW, Australia
| | - David Espinoza
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - John P Grady
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Chee Khoon Lee
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Mandy L Ballinger
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Lucille Sebastian
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Theresa Corpuz
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Min Ru Qiu
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Department of Anatomical Pathology and Cancer Genetics, SydPath, St Vincent's Hospital, Sydney, NSW, Australia
| | - Piyushkumar Mundra
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Charles G Bailey
- Cancer & Gene Regulation Laboratory Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Gene and Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia
| | - Ulf Schmitz
- Gene and Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia
- Computational Biomedicine Lab Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
- Department of Molecular & Cell Biology, College of Public Health, Medical & Vet Sciences, James Cook University, Townsville, QLD, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Anthony M Joshua
- The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - David M Thomas
- The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Biomedical Science, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
8
|
Rozeman EA, Versluis JM, Sikorska K, Hoefsmit EP, Dimitriadis P, Rao D, Lacroix R, Grijpink-Ongering LG, Lopez-Yurda M, Heeres BC, van de Wiel BA, Flohil C, Sari A, Heijmink SWTPJ, van den Broek D, Broeks A, de Groot JWB, Vollebergh MA, Wilgenhof S, van Thienen JV, Haanen JBAG, Blank CU. IMPemBra: a phase 2 study comparing pembrolizumab with intermittent/short-term dual MAPK pathway inhibition plus pembrolizumab in patients with melanoma harboring the BRAFV600 mutation. J Immunother Cancer 2023; 11:e006821. [PMID: 37479483 PMCID: PMC10364170 DOI: 10.1136/jitc-2023-006821] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Continuous combination of MAPK pathway inhibition (MAPKi) and anti-programmed death-(ligand) 1 (PD-(L)1) showed high response rates, but only limited improvement in progression-free survival (PFS) at the cost of a high frequency of treatment-related adverse events (TRAE) in patients with BRAFV600-mutated melanoma. Short-term MAPKi induces T-cell infiltration in patients and is synergistic with anti-programmed death-1 (PD-1) in a preclinical melanoma mouse model. The aim of this phase 2b trial was to identify an optimal regimen of short-term MAPKi with dabrafenib plus trametinib in combination with pembrolizumab. METHODS Patients with treatment-naïve BRAFV600E/K-mutant advanced melanoma started pembrolizumab 200 mg every 3 weeks. In week 6, patients were randomized to continue pembrolizumab only (cohort 1), or to receive, in addition, intermittent dabrafenib 150 mg two times per day plus trametinib 2 mg one time per day for two cycles of 1 week (cohort 2), two cycles of 2 weeks (cohort 3), or continuously for 6 weeks (cohort 4). All cohorts continued pembrolizumab for up to 2 years. Primary endpoints were safety and treatment-adherence. Secondary endpoints were objective response rate (ORR) at week 6, 12, 18 and PFS. RESULTS Between June 2016 and August 2018, 33 patients with advanced melanoma have been included and 32 were randomized. Grade 3-4 TRAE were observed in 12%, 12%, 50%, and 63% of patients in cohort 1, 2, 3, and 4, respectively. All planned targeted therapy was given in 88%, 63%, and 38% of patients in cohort 2, 3, and 4. ORR at week 6, 12, and 18 were 38%, 63%, and 63% in cohort 1; 25%, 63%, and 75% in cohort 2; 25%, 50%, and 75% in cohort 3; and 0%, 63%, and 50% in cohort 4. After a median follow-up of 43.5 months, median PFS was 10.6 months for pembrolizumab monotherapy and not reached for patients treated with pembrolizumab and intermittent dabrafenib and trametinib (p=0.17). The 2-year and 3-year landmark PFS were both 25% for cohort 1, both 63% for cohort 2, 50% and 38% for cohort 3 and 75% and 60% for cohort 4. CONCLUSIONS The combination of pembrolizumab plus intermittent dabrafenib and trametinib seems more feasible and tolerable than continuous triple therapy. The efficacy is promising and appears to be favorable over pembrolizumab monotherapy. TRIAL REGISTRATION NUMBER NCT02625337.
Collapse
Affiliation(s)
- Elisa A Rozeman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Judith M Versluis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Karolina Sikorska
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esmée P Hoefsmit
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petros Dimitriadis
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Disha Rao
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ruben Lacroix
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marta Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Birthe C Heeres
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bart A van de Wiel
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Claudie Flohil
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Aysegul Sari
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Daan van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Annegien Broeks
- Core Facility and Biobanking, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marieke A Vollebergh
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Johannes V van Thienen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical center, Leiden, The Netherlands
| |
Collapse
|
9
|
Cazzato G, Lettini T, Colagrande A, Trilli I, Ambrogio F, Laface C, Parente P, Maiorano E, Ingravallo G. Immunohistochemical Expression of Programmed Cell Death Ligand 1 (PD-L1) in Human Cutaneous Malignant Melanoma: A Narrative Review with Historical Perspectives. Genes (Basel) 2023; 14:1252. [PMID: 37372432 DOI: 10.3390/genes14061252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is the primary ligand of the receptor programmed death-1 (PD-1) which is constitutively expressed or activated in myeloid, lymphoid (T, B and NK), normal epithelial cells, and cancer. The PD-1/PD-L1 interaction is crucial for the physiological development of immunological tolerance but also in the development of the cancer. Among these, malignant melanoma represents a tumour in which the immunohistochemical expression of PD-L1 is important to guide future therapeutic choices based on the presence/absence of expression. Various clones have been used over time for immunohistochemical determination, and different results and heterogeneity remain among the various studies in the literature. We perform a narrative review of the present studies in order to discuss and take stock of what certain achievements have been made in this field, what challenges remain, and what possible solutions can be found.
Collapse
Affiliation(s)
- Gerardo Cazzato
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Teresa Lettini
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Anna Colagrande
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Irma Trilli
- Odontomatostologic Clinic, Department of Innovative Technologies in Medicine and Dentistry, University of Chieti, 66100 Chieti, Italy
| | - Francesca Ambrogio
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, BR, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy
| | - Eugenio Maiorano
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Giuseppe Ingravallo
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| |
Collapse
|
10
|
Haist M, Stege H, Kuske M, Bauer J, Klumpp A, Grabbe S, Bros M. Combination of immune-checkpoint inhibitors and targeted therapies for melanoma therapy: The more, the better? Cancer Metastasis Rev 2023; 42:481-505. [PMID: 37022618 PMCID: PMC10348973 DOI: 10.1007/s10555-023-10097-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 04/07/2023]
Abstract
The approval of immune-checkpoint inhibitors (CPI) and mitogen activated protein kinase inhibitors (MAPKi) in recent years significantly improved the treatment management and survival of patients with advanced malignant melanoma. CPI aim to counter-act receptor-mediated inhibitory effects of tumor cells and immunomodulatory cell types on effector T cells, whereas MAPKi are intended to inhibit tumor cell survival. In agreement with these complementary modes of action preclinical data indicated that the combined application of CPI and MAPKi or their optimal sequencing might provide additional clinical benefit. In this review the rationale and preclinical evidence that support the combined application of MAPKi and CPI either in concurrent or consecutive regimens are presented. Further, we will discuss the results from clinical trials investigating the sequential or combined application of MAPKi and CPI for advanced melanoma patients and their implications for clinical practice. Finally, we outline mechanisms of MAPKi and CPI cross-resistance which limit the efficacy of currently available treatments, as well as combination regimens.
Collapse
Affiliation(s)
- Maximilian Haist
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Michael Kuske
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Julia Bauer
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Annika Klumpp
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
11
|
Derks SHAE, Jongen JLM, van der Meer EL, Ho LS, Slagter C, Joosse A, de Jonge MJA, Schouten JW, Oomen-de Hoop E, van den Bent MJ, van der Veldt AAM. Impact of Novel Treatments in Patients with Melanoma Brain Metastasis: Real-World Data. Cancers (Basel) 2023; 15:cancers15051461. [PMID: 36900253 PMCID: PMC10000692 DOI: 10.3390/cancers15051461] [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: 02/01/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Melanoma brain metastasis (MBM) is associated with poor outcome, but targeted therapies (TTs) and immune checkpoint inhibitors (ICIs) have revolutionized treatment over the past decade. We assessed the impact of these treatments in a real-world setting. METHODS A single-center cohort study was performed at a large, tertiary referral center for melanoma (Erasmus MC, Rotterdam, the Netherlands). Overall survival (OS) was assessed before and after 2015, after which TTs and ICIs were increasingly prescribed. RESULTS There were 430 patients with MBM included; 152 pre-2015 and 278 post-2015. Median OS improved from 4.4 to 6.9 months (HR 0.67, p < 0.001) after 2015. TTs and ICIs prior to MBM diagnosis were associated with poorer median OS as compared to no prior systemic treatment (TTs: 2.0 vs. 10.9 and ICIs: 4.2 vs. 7.9 months, p < 0.001). ICIs directly after MBM diagnosis were associated with improved median OS as compared to no direct ICIs (21.5 vs. 4.2 months, p < 0.001). Stereotactic radiotherapy (SRT; HR 0.49, p = 0.013) and ICIs (HR 0.32, p < 0.001) were independently associated with improved OS. CONCLUSION After 2015, OS significantly improved for patients with MBM, especially with SRT and ICIs. Demonstrating a large survival benefit, ICIs should be considered first after MBM diagnosis, if clinically feasible.
Collapse
Affiliation(s)
- Sophie H. A. E. Derks
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Joost L. M. Jongen
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Edgar L. van der Meer
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Li Shen Ho
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Cleo Slagter
- Department of Radiotherapy, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Maja J. A. de Jonge
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Joost W. Schouten
- Department of Neurosurgery, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Martin J. van den Bent
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Astrid A. M. van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Correspondence: ; Tel.: +31-10-704-02-52
| |
Collapse
|
12
|
Barnestein R, Galland L, Kalfeist L, Ghiringhelli F, Ladoire S, Limagne E. Immunosuppressive tumor microenvironment modulation by chemotherapies and targeted therapies to enhance immunotherapy effectiveness. Oncoimmunology 2022; 11:2120676. [PMID: 36117524 PMCID: PMC9481153 DOI: 10.1080/2162402x.2022.2120676] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
With the rapid clinical development of immune checkpoint inhibitors (ICIs), the standard of care in cancer management has evolved rapidly. However, immunotherapy is not currently beneficial for all patients. In addition to intrinsic tumor factors, other etiologies of resistance to ICIs arise from the complex interplay between cancer and its microenvironment. Recognition of the essential role of the tumor microenvironment (TME) in cancer progression has led to a shift from a tumor-cell-centered view of cancer development, to the concept of a complex tumor ecosystem that supports tumor growth and metastatic dissemination. The expansion of immunosuppressive cells represents a cardinal strategy deployed by tumor cells to escape detection and elimination by the immune system. Regulatory T lymphocytes (Treg), myeloid-derived suppressor cells (MDSCs), and type-2 tumor-associated macrophages (TAM2) are major components of these inhibitory cellular networks, with the ability to suppress innate and adaptive anticancer immunity. They therefore represent major impediments to anticancer therapies, particularly immune-based interventions. Recent work has provided evidence that, beyond their direct cytotoxic effects on cancer cells, several conventional chemotherapeutic (CT) drugs and agents used in targeted therapies (TT) can promote the elimination or inactivation of suppressive immune cells, resulting in enhanced antitumor immunity. In this review, we will analyze findings pertaining to this concept, discuss the possible molecular bases underlying the selective targeting of these immunosuppressive cells by antineoplastic agents (CT and/or TT), and consider current challenges and future prospects related to the integration of these molecules into more efficient anticancer strategies, in the era of immunotherapy.
Collapse
Affiliation(s)
- Robby Barnestein
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
| | - Loïck Galland
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
| | - Laura Kalfeist
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - François Ghiringhelli
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - Sylvain Ladoire
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - Emeric Limagne
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| |
Collapse
|
13
|
Caksa S, Baqai U, Aplin AE. The future of targeted kinase inhibitors in melanoma. Pharmacol Ther 2022; 239:108200. [PMID: 35513054 PMCID: PMC10187889 DOI: 10.1016/j.pharmthera.2022.108200] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022]
Abstract
Melanoma is a cancer of the pigment-producing cells of the body and its incidence is rising. Targeted inhibitors that act against kinases in the MAPK pathway are approved for BRAF-mutant metastatic cutaneous melanoma and increase patients' survival. Response to these therapies is limited by drug resistance and is less durable than with immune checkpoint inhibition. Conversely, rare melanoma subtypes have few therapeutic options for advanced disease and MAPK pathway targeting agents show minimal anti-tumor effects. Nevertheless, there is a future for targeted kinase inhibitors in melanoma: in new applications such as adjuvant or neoadjuvant therapy and in novel combinations with immunotherapies or other targeted therapies. Pre-clinical studies continue to identify tumor dependencies and their corresponding actionable drug targets, paving the way for rational targeted kinase inhibitor combinations as a personalized medicine approach for melanoma.
Collapse
Affiliation(s)
- Signe Caksa
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Usman Baqai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
14
|
Bai X, Quek C. Unravelling Tumour Microenvironment in Melanoma at Single-Cell Level and Challenges to Checkpoint Immunotherapy. Genes (Basel) 2022; 13:genes13101757. [PMID: 36292642 PMCID: PMC9601741 DOI: 10.3390/genes13101757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Melanoma is known as one of the most immunogenic tumours and is often characterised by high mutation burden, neoantigen load and immune infiltrate. The application of immunotherapies has led to impressive improvements in the clinical outcomes of advanced stage melanoma patients. The standard of care immunotherapies leverage the host immunological influence on tumour cells, which entail complex interactions among the tumour, stroma, and immune cells at the tumour microenvironmental level. However, not all cancer patients can achieve a long-term durable response to immunotherapy, and a significant proportion of patients develops resistance and still die from their disease. Owing to the multi-faceted problems of tumour and microenvironmental heterogeneity, identifying the key factors underlying tumour progression and immunotherapy resistance poses a great challenge. In this review, we outline the main challenges to current cancer immunotherapy research posed by tumour heterogeneity and microenvironment complexities including genomic and transcriptomic variability, selective outgrowth of tumour subpopulations, spatial and temporal tumour heterogeneity and the dynamic state of host immunity and microenvironment orchestration. We also highlight the opportunities to dissect tumour heterogeneity using single-cell sequencing and spatial platforms. Integrative analyses of large-scale datasets will enable in-depth exploration of biological questions, which facilitates the clinical application of translational research.
Collapse
|
15
|
Gritsch D, Mrugala MM, Marks LA, Wingerchuk DM, O'Carroll CB. In Patients With Melanoma Brain Metastases, Is Combination Immune Checkpoint Inhibition a Safe and Effective First-Line Treatment? A Critically Appraised Topic. Neurologist 2022; 27:290-297. [PMID: 35834790 DOI: 10.1097/nrl.0000000000000439] [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: 11/26/2022]
Abstract
BACKGROUND Combined PD-1/PD-L1 and CTLA-4 immune checkpoint inhibition for the has been shown to produce superior results in the treatment of malignant melanoma when compared to monotherapy. However, patients with intracranial disease were excluded from these studies given their poor prognosis. OBJECTIVE The objective of this study was to critically assess current evidence supporting the co-administration of PD-1/PD-L1 and CTLA-4 inhibitors in the treatment of melanoma brain metastases. METHODS The objective was addressed through the development of a critically appraised topic that included a clinical scenario, structured question, literature search strategy, critical appraisal, assessment of results, evidence summary, commentary, and bottom-line conclusions. Participants included consultant and resident neurologists, a medical librarian, clinical epidemiologists, and a content expert in the field of neuro-oncology. RESULTS A recent, open-label, non-comparative randomized phase II trial was selected for critical appraisal. This trial evaluated the efficacy and safety of nivolumab alone or in combination with ipilimumab in 79 adult patients with untreated, asymptomatic melanoma brain metastases. The rates of the primary outcome (intracranial response at ≥12 wk) in the primary endpoint cohort were 46% for cohort A (combination therapy) and 20% for cohort B (nivolumab monotherapy). No treatment related deaths were observed in the study. Grade 4 adverse events occurred in 9% of patients in cohort A and none in cohort B. CONCLUSIONS Co-administration of ipilimumab and nivolumab as first-line therapy is effective in the treatment of asymptomatic melanoma brain metastases, with an acceptable safety profile.
Collapse
|
16
|
Zhang Z, Richmond A, Yan C. Immunomodulatory Properties of PI3K/AKT/mTOR and MAPK/MEK/ERK Inhibition Augment Response to Immune Checkpoint Blockade in Melanoma and Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms23137353. [PMID: 35806358 PMCID: PMC9266842 DOI: 10.3390/ijms23137353] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 02/03/2023] Open
Abstract
Hyperactivation of PI3K/AKT/mTOR and MAPK/MEK/ERK signaling pathways is commonly observed in many cancers, including triple-negative breast cancer (TNBC) and melanoma. Moreover, the compensatory upregulation of the MAPK/MEK/ERK pathway has been associated with therapeutic resistance to targeted inhibition of the PI3K/AKT/mTOR pathway, and vice versa. The immune-modulatory effects of both PI3K and MAPK inhibition suggest that inhibition of these pathways might enhance response to immune checkpoint inhibitors (ICIs). ICIs have become the standard-of-care for metastatic melanoma and are recently an option for TNBC when combined with chemotherapy, but alternative options are needed when resistance develops. In this review, we present the current mechanistic understandings, along with preclinical and clinical evidence, that outline the efficacy and safety profile of combinatorial or sequential treatments with PI3K inhibitors, MAPK inhibitors, and ICIs for treatment of malignant melanoma and metastatic TNBC. This approach may present a potential strategy to overcome resistance in patients who are a candidate for ICI therapy with tumors harboring either or both of these pathway-associated mutations.
Collapse
Affiliation(s)
- Zhizhu Zhang
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; (Z.Z.); (A.R.)
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; (Z.Z.); (A.R.)
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Chi Yan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; (Z.Z.); (A.R.)
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
- Correspondence:
| |
Collapse
|
17
|
Eggermont AMM, Hamid O, Long GV, Luke JJ. Optimal systemic therapy for high-risk resectable melanoma. Nat Rev Clin Oncol 2022; 19:431-439. [PMID: 35468949 PMCID: PMC11075933 DOI: 10.1038/s41571-022-00630-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
Abstract
Immunotherapy with immune-checkpoint inhibitors and molecularly targeted therapy with BRAF inhibitors were pioneered in the setting of advanced-stage, unresectable melanoma, where they revolutionized treatment and considerably improved patient survival. These therapeutic approaches have also been successfully transitioned into the resectable disease setting, with the regulatory approvals of ipilimumab, pembrolizumab, nivolumab, and dabrafenib plus trametinib as postoperative (adjuvant) treatments for various, overlapping groups of patients with high-risk melanoma. Moreover, these agents have shown variable promise when used in the preoperative (neoadjuvant) period. The expanding range of treatment options available for resectable high-risk melanoma, all of which come with risks as well as benefits, raises questions over selection of the optimal therapeutic strategy and agents for each individual, also considering that many patients might be cured with surgery alone. Furthermore, the use of perioperative therapy has potentially important implications for the management of patients who have disease recurrence. In this Viewpoint, we asked four expert investigators and medical or surgical oncologists who have been involved in the key studies of perioperative systemic therapies for their perspectives on the optimal management of patients with high-risk melanoma.
Collapse
Affiliation(s)
- Alexander M M Eggermont
- Comprehensive Cancer Center Munich, Munich, Germany.
- Princess Máxima Center for Paediatric Oncology, Utrecht, Netherlands.
- University Medical Center Utrecht, Utrecht, Netherlands.
| | - Omid Hamid
- The Angeles Clinic and Research Institute, Cedar Sinai Affiliate, Los Angeles, CA, USA.
| | - Georgia V Long
- Melanoma Institute Australia and Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
- Mater and Royal North Shore Hospitals, Sydney, New South Wales, Australia.
| | - Jason J Luke
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Pires da Silva I, Zakria D, Ahmed T, Trojanello C, Dimitriou F, Allayous C, Gerard C, Zimmer L, Lo S, Michielin O, Lebbe C, Mangana J, Ascierto PA, Johnson DB, Carlino M, Menzies A, Long G. Efficacy and safety of anti-PD1 monotherapy or in combination with ipilimumab after BRAF/MEK inhibitors in patients with BRAF mutant metastatic melanoma. J Immunother Cancer 2022; 10:e004610. [PMID: 35798536 PMCID: PMC9263926 DOI: 10.1136/jitc-2022-004610] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Patients with V600BRAF mutant metastatic melanoma have higher rates of progression-free survival (PFS) and overall survival (OS) with first-line anti-PD1 (PD1]+anti-CTLA-4 (IPI) versus PD1. Whether this is also true after BRAF/MEKi therapy is unknown. We aimed to determine the efficacy and safety of PD1 versus IPI +PD1 after BRAF/MEK inhibitors (BRAF/MEKi). METHODS Patients with V600BRAF mutant metastatic melanoma treated with BRAF/MEKi who had subsequent PD1 versus IPI+PD1 at eight centers were included. The endpoints were objective response rate (ORR), PFS, OS and safety in each group. RESULTS Of 200 patients with V600E (75%) or non-V600E (25%) mutant metastatic melanoma treated with BRAF/MEKi (median time of treatment 7.6 months; treatment cessation due to progressive disease in 77%), 115 (57.5%) had subsequent PD1 and 85 (42.5%) had IPI+PD1. Differences in patient characteristics between PD1 and IPI+PD1 groups included, age (med. 63 vs 54 years), time between BRAF/MEKi and PD1±IPI (16 vs 4 days), Eastern Cooperative Oncology Group Performance Status (ECOG PS) of ≥1 (62% vs 44%), AJCC M1C/M1D stage (72% vs 94%) and progressing brain metastases at the start of PD1±IPI (34% vs 57%). Median follow-up from PD1±IPI start was 37.8 months (95% CI, 33.9 to 52.9). ORR was 36%; 34% with PD1 vs 39% with IPI+PD1 (p=0.5713). Median PFS was 3.4 months; 3.4 with PD1 vs 3.6 months with IPI+PD1 (p=0.6951). Median OS was 15.4 months; 14.4 for PD1 vs 20.5 months with IPI+PD1 (p=0.2603). The rate of grade 3 or 4 toxicities was higher with IPI+PD1 (31%) vs PD1 (7%). ORR, PFS and OS were numerically higher with IPI+PD1 vs PD1 across most subgroups except for females, those with <10 days between BRAF/MEKi and PD1±IPI, and those with stage III/M1A/M1B melanoma. The combination of ECOG PS=0 and absence of liver metastases identified patients with >3 years OS (area under the curve, AUC=0.74), while ECOG PS ≥1, progressing brain metastases and presence of bone metastases predicted primary progression (AUC=0.67). CONCLUSIONS IPI+PD1 and PD1 after BRAF/MEKi have similar outcomes despite worse baseline prognostic features in the IPI+PD1 group, however, IPI+PD1 is more toxic. A combination of clinical factors can identify long-term survivors, but less accurately those with primary resistance to immunotherapy after targeted therapy.
Collapse
Affiliation(s)
- Ines Pires da Silva
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Blacktown & Westmead Hospital, Sydney, New South Wales, Australia
| | - Danny Zakria
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tasnia Ahmed
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia
| | - Claudia Trojanello
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Clara Allayous
- Dermatolo-Oncology AP-HP Hôpital Saint-Louis, INSERM U976, Paris, France
| | | | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Serigne Lo
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia
| | | | - Celeste Lebbe
- Université Paris Cite, Dermatolo-Oncology AP-HP Hôpital Saint-Louis, INSERM U976, Paris, France
| | - Johanna Mangana
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | - Matteo Carlino
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia
- Blacktown & Westmead Hospital, Sydney, New South Wales, Australia
| | - Alexander Menzies
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Georgina Long
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
Schadendorf D, Dummer R, Robert C, Ribas A, Sullivan RJ, Panella T, McKean M, Santos ES, Brill K, Polli A, Pietro AD, Ascierto PA. STARBOARD: encorafenib + binimetinib + pembrolizumab for first-line metastatic/unresectable BRAF V600-mutant melanoma. Future Oncol 2022; 18:2041-2051. [PMID: 35272485 DOI: 10.2217/fon-2021-1486] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite the significant progress in the treatment of unresectable or metastatic BRAF V600-mutant melanoma, there remains two primary treatment options: targeted therapy and immunotherapy. Targeted therapy or immunotherapy alone is associated with efficacy limitations including efficacy limited to select patient subsets. With separate mechanisms of action and different response patterns, the combination of targeted agents and immunotherapy to treat patients with BRAF V600-mutant melanoma may further improve patient outcomes. Current treatment guidelines recommend treatment with targeted agents alone, immunotherapy, or the combination of targeted agents and immunotherapy. The randomized, double-blind STARBOARD trial aims to evaluate efficacy and safety of encorafenib, binimetinib and pembrolizumab in treatment-naive patients with metastatic or unresectable locally advanced BRAF V600-mutant melanoma in comparison to pembrolizumab.
Collapse
Affiliation(s)
- Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, 45122 Essen, Germany & German Cancer Consortium, Partner Site Essen, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zürich & University Zurich, Zurich, 8091, Switzerland
| | - Caroline Robert
- Melanoma Research Unit, Institut Gustave Roussy, Villejuif, 94805, France
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ryan J Sullivan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Timothy Panella
- Department of Medicine, Division of Hematology & Oncology, University of Tennessee, Knoxville, TN 37996, USA
| | - Meredith McKean
- Sarah Cannon Research Institute at Tennessee Oncology, Nashville, TN 37203, USA
| | - Edgardo S Santos
- Florida Precision Oncology/A Division of Genesis Care USA, Florida Atlantic University, Aventura, FL 33180, USA
| | | | | | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, 80131, Italy
| |
Collapse
|
20
|
Bari S, Muzaffar J, Eroglu Z. Combination targeted and immune therapy in the treatment of advanced melanoma: a valid treatment option for patients? Ther Adv Med Oncol 2022; 14:17588359221090306. [PMID: 35478991 PMCID: PMC9036333 DOI: 10.1177/17588359221090306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Melanomas harboring an activating BRAFV600 mutation account for 50% of all advanced melanomas. The approval of BRAF-targeted therapy revolutionized treatment of these patients with achievement of impressive responses. However, development of resistance to these drugs is a significant problem, and as such, duration of response remains low, with median progression free survival of around 11–15 months. Immune checkpoint blockers exploit the immune system to eradicate cancer and can produce durable disease control that results in long-term, treatment-free survival in some patients. These drugs have shown very impressive survival in patients with BRAF-mutated melanoma. Thus, there is a need to continue to utilize emerging data to achieve long-term disease control for patients with advanced melanoma. Combining targeted therapy with immune therapy may be one possible way to achieve this goal. In this review, the mechanisms of action of these two pathways, including the mechanistic basis of this combination, are summarized, along with results of completed and ongoing trials in triple therapy.
Collapse
Affiliation(s)
- Shahla Bari
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jameel Muzaffar
- Department of Head and Neck Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
21
|
Dixon-Douglas JR, Patel RP, Somasundram PM, McArthur GA. Triplet Therapy in Melanoma - Combined BRAF/MEK Inhibitors and Anti-PD-(L)1 Antibodies. Curr Oncol Rep 2022; 24:1071-1079. [PMID: 35366166 PMCID: PMC9249697 DOI: 10.1007/s11912-022-01243-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2021] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW We provide an updated review of clinical trials evaluating the combination of BRAF/MEK inhibitors with anti-PD-(L)1 therapy (triplet therapy) for patients with advanced BRAF-mutant melanoma, accompanied by a summary of the biological evidence supporting this combination. RECENT FINDINGS Resistance to BRAF/MEK inhibition and comparatively low response rates to immune checkpoint inhibitors remain clinical challenges in the treatment of melanoma. Preclinical data demonstrates that targeted therapy is immune-modulatory and synergises with immune checkpoint inhibition. Several randomised controlled trials have evaluated the combination of targeted therapy with immune checkpoint inhibition. Triplet therapy has shown improvements in progression-free survival and durability of response compared to BRAF/MEK inhibition alone; however, questions remain regarding the best clinical scenario for implementation of this regimen in the era of front-line immunotherapy.
Collapse
Affiliation(s)
- Julia R Dixon-Douglas
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Riyaben P Patel
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Pretashini M Somasundram
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Grant A McArthur
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Department of Oncology, Sir Peter MacCallum, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
22
|
Morrison SL, Han G, Elenwa F, Vetto JT, Fowler G, Leong SP, Kashani-Sabet M, Pockaj BA, Kosiorek HE, Zager JS, Sondak VK, Messina JL, Mozzillo N, Schneebaum S, Han D. Is the presence of tumor-infiltrating lymphocytes predictive of outcomes in patients with melanoma? Cancer 2022; 128:1418-1428. [PMID: 35103302 DOI: 10.1002/cncr.34013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The significance of tumor-infiltrating lymphocytes (TILs) in melanoma is debated. This article presents a multicenter, retrospective study assessing the predictive and prognostic value of TILs. METHODS The Sentinel Lymph Node Working Group database was queried from 1993 to 2018 for cases with known TIL data. TILs were categorized as absent or present, which included nonbrisk (NB), brisk (B), and present but unspecified TIL levels. Clinicopathologic factors were correlated with TILs, sentinel lymph node (SLN) status, and melanoma-specific survival (MSS). RESULTS Overall, 3203 patients were included. The median thickness was 1.5 mm, and 469 cases had SLN metastases. TILs were present in 2458 cases (76.7%), with NB, B, and unspecified TILs seen in 1691 (68.8%), 691 (28.1%), and 76 (3.1%), respectively. Multivariable analysis showed that the presence of TILs significantly predicted a negative SLN biopsy (P < .05). The median follow-up was 25.2 months. MSS was significantly better for cases with TILs than cases without TILs (P < .001). According to multivariable analysis, age, gender, thickness, mitotic rate, ulceration, lymphovascular invasion, and SLN status were significantly prognostic of MSS (all P values < .05). Although TILs were not prognostic of MSS, when multiple imputation was used and the SLN status was excluded, the presence of TILs was significantly prognostic of improved MSS (hazard ratio, 0.78; 95% confidence interval, 0.64-0.95; P = .0154). CONCLUSIONS TILs are a favorable marker because their presence significantly predicts a negative SLN, and the absence of TILs may be a prognostic marker of worse survival in patients with a positive SLN but not a negative SLN. TILs may also serve as a prognostic marker of survival when the SLN status is not considered.
Collapse
Affiliation(s)
- Steven L Morrison
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Gang Han
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas
| | - Faith Elenwa
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas
| | - John T Vetto
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Graham Fowler
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Stanley P Leong
- California Pacific Medical Center and Research Institute, San Francisco, California
| | | | | | | | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Jane L Messina
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | | | - Dale Han
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
23
|
Newell F, Pires da Silva I, Johansson PA, Menzies AM, Wilmott JS, Addala V, Carlino MS, Rizos H, Nones K, Edwards JJ, Lakis V, Kazakoff SH, Mukhopadhyay P, Ferguson PM, Leonard C, Koufariotis LT, Wood S, Blank CU, Thompson JF, Spillane AJ, Saw RPM, Shannon KF, Pearson JV, Mann GJ, Hayward NK, Scolyer RA, Waddell N, Long GV. Multiomic profiling of checkpoint inhibitor-treated melanoma: Identifying predictors of response and resistance, and markers of biological discordance. Cancer Cell 2022; 40:88-102.e7. [PMID: 34951955 DOI: 10.1016/j.ccell.2021.11.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 09/15/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
We concurrently examine the whole genome, transcriptome, methylome, and immune cell infiltrates in baseline tumors from 77 patients with advanced cutaneous melanoma treated with anti-PD-1 with or without anti-CTLA-4. We show that high tumor mutation burden (TMB), neoantigen load, expression of IFNγ-related genes, programmed death ligand expression, low PSMB8 methylation (therefore high expression), and T cells in the tumor microenvironment are associated with response to immunotherapy. No specific mutation correlates with therapy response. A multivariable model combining the TMB and IFNγ-related gene expression robustly predicts response (89% sensitivity, 53% specificity, area under the curve [AUC], 0.84); tumors with high TMB and a high IFNγ signature show the best response to immunotherapy. This model validates in an independent cohort (80% sensitivity, 59% specificity, AUC, 0.79). Except for a JAK3 loss-of-function mutation, for patients who did not respond as predicted there is no obvious biological mechanism that clearly explained their outlier status, consistent with intratumor and intertumor heterogeneity in response to immunotherapy.
Collapse
Affiliation(s)
- Felicity Newell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Ines Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Cancer Centre, Blacktown Hospital, Sydney, NSW 2148, Australia
| | - Peter A Johansson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW 2065, Australia; Mater Hospital, Sydney, NSW 2060, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Venkateswar Addala
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Centre for Cancer Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia; Department of Medical Oncology, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Helen Rizos
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, NSW 2109, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Jarem J Edwards
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Vanessa Lakis
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Stephen H Kazakoff
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | | | - Peter M Ferguson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Camperdown, NSW 2050, Australia
| | - Conrad Leonard
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | | | - Scott Wood
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Christian U Blank
- Department of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Mater Hospital, Sydney, NSW 2060, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Andrew J Spillane
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Mater Hospital, Sydney, NSW 2060, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Mater Hospital, Sydney, NSW 2060, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Kerwin F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Mater Hospital, Sydney, NSW 2060, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - John V Pearson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Graham J Mann
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Centre for Cancer Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia; John Curtin School of Medical Research, Australian National University, ACT 2601, Australia
| | - Nicholas K Hayward
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Camperdown, NSW 2050, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW 2065, Australia; Mater Hospital, Sydney, NSW 2060, Australia.
| |
Collapse
|
24
|
Chen Y, Feng R, He B, Wang J, Xian N, Huang G, Zhang Q. PD-1H Expression Associated With CD68 Macrophage Marker Confers an Immune-Activated Microenvironment and Favorable Overall Survival in Human Esophageal Squamous Cell Carcinoma. Front Mol Biosci 2021; 8:777370. [PMID: 34950702 PMCID: PMC8688962 DOI: 10.3389/fmolb.2021.777370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal carcinoma (EC) in China. Although the PD-1 inhibitor pembrolizumab has been approved to treat patients with EC, its therapeutic efficacy is limited. Thus, additional immunotherapeutic targets for EC treatment are needed. Programmed Death-1 Homolog (PD-1H) is a negative checkpoint regulator that inhibits antitumor immune responses. Here, PD-1H expression in 114 patients with ESCC was evaluated by immunohistochemistry. Next, 12 randomly selected tumor tissue sections were used to assess the colocalization of PD-1H protein and multiple immune markers by multiplex immunohistochemistry. Our results demonstrated that PD-1H was expressed at high frequency in ESCC tumor tissues (85.1%). PD-1H protein was predominantly expressed in CD68+ tumor-associated macrophages and expressed at low levels in CD4+ T cells and CD8+ T cells in ESCC tumor tissues. Furthermore, based on ESCC data in The Cancer Genome Atlas (TCGA), the gene expression levels of PD-1H were positively associated with the infiltration levels of immune-activated cells especially CD8+ cytotoxic T cells. In contrast, the gene expression levels of PD-1H were negatively correlated with myeloid-derived suppressor cells (MDSCs). Importantly, PD-1H expression in tumor sites was significantly correlated with favorable overall survival in patients with ESCC. Collectively, our findings first provided direct information on the PD-1H expression pattern and distribution in ESCC, and positive correlation of PD-1H expression with overall survival suggested PD-1H expression levels could be a significant prognostic indicator for patients with ESCC. Future studies need to explore the immunoregulatory of PD-1H in the tumor microenvironment of ESCC.
Collapse
Affiliation(s)
- Yuangui Chen
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui Feng
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.,Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bailin He
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jun Wang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Na Xian
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Gangxiong Huang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Qiuyu Zhang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
25
|
Long GV, Arance A, Mortier L, Lorigan P, Blank C, Mohr P, Schachter J, Grob JJ, Lotem M, Middleton MR, Neyns B, Steven N, Ribas A, Walpole E, Carlino MS, Lebbe C, Sznol M, Jensen E, Leiby MA, Ibrahim N, Robert C. Antitumor activity of ipilimumab or BRAF ± MEK inhibition after pembrolizumab treatment in patients with advanced melanoma: analysis from KEYNOTE-006. Ann Oncol 2021; 33:204-215. [PMID: 34710571 DOI: 10.1016/j.annonc.2021.10.010] [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: 06/29/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Antitumor activity of ipilimumab or BRAF ± MEK inhibitors (BRAFi ± MEKi) following pembrolizumab administration in melanoma is poorly characterized. PATIENTS AND METHODS In the phase III KEYNOTE-006 study, patients with unresectable stage III/IV melanoma received pembrolizumab (10 mg/kg) once every 2 or 3 weeks (Q3W) or ipilimumab (3 mg/kg) Q3W. The current post hoc analysis evaluates outcomes with ipilimumab or BRAFi ± MEKi as first subsequent systemic therapy after pembrolizumab administration and includes patients who completed or discontinued pembrolizumab after one or more dose. Pembrolizumab arms were pooled. RESULTS At data cut-off (4 December 2017), median follow-up was 46.9 months. Of 555 pembrolizumab-treated patients, first subsequent therapy was ipilimumab for 103 (18.6%) and BRAFi ± MEKi for 59 (10.6%) [33 received BRAFi + MEKi, 26 BRAFi alone; 37 (62.7%) were BRAFi ± MEKi naïve]. In the subsequent ipilimumab group, ORR with previous pembrolizumab was 17.5% [1 complete response (CR); 17 partial response (PR)]; 79.6% had discontinued pembrolizumab due to progressive disease (PD); median overall survival (OS) was 21.5 months. ORR with subsequent ipilimumab was 15.5%; 11/16 responses (8 CRs; 3 PRs) were ongoing. ORR with subsequent ipilimumab was 9.7% for patients with PD as best response to pembrolizumab. Median OS from ipilimumab initiation was 9.8 months. In the subsequent BRAFi ± MEKi group, ORR with previous pembrolizumab was 13.5% (8 PR); 76.3% had discontinued pembrolizumab due to PD; median OS was 17.9 months. ORR with subsequent BRAFi ± MEKi was 30.5%, 7/18 responses (4 CR, 3 PR) were ongoing. Median OS from BRAFi ± MEKi initiation was 12.9 months. ORR for BRAFi ± MEKi-naïve patients who received subsequent BRAFi ± MEKi was 43.2%; 6/16 were ongoing (3 CR, 3 PR). CONCLUSIONS Ipilimumab and BRAFi ± MEKi have antitumor activity as first subsequent therapy after pembrolizumab in patients with advanced melanoma.
Collapse
Affiliation(s)
- G V Long
- Melanoma Institute Australia, The University of Sydney, Mater Hospital, Sydney, Australia; Faculty of Medicine & Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia; Royal North Shore Hospital, Sydney, Australia.
| | - A Arance
- Hospital Clinic de Barcelona, Barcelona, Spain
| | - L Mortier
- Université Lille, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - P Lorigan
- Division of Cancer Sciences, University of Manchester, Manchester; Christie NHS Foundation Trust, Manchester, UK
| | - C Blank
- Netherlands Cancer Institute, Amsterdam, Netherlands
| | - P Mohr
- Elbe-Klinikum Buxtehude, Buxtehude, Germany
| | - J Schachter
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center at Tel Hashomer, Ramat Gan, Israel
| | - J-J Grob
- Aix Marseille University, Hôpital de la Timone, Marseille, France
| | - M Lotem
- Sharett Institute of Oncology, Hadassah Hebrew Medical Center, Jerusalem, Israel
| | - M R Middleton
- The Churchill Hospital and The University of Oxford, Oxford, UK
| | - B Neyns
- Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - N Steven
- Queen Elizabeth Hospital, Birmingham, UK
| | - A Ribas
- David Geffen School of Medicine, University of California, Los Angeles, USA
| | - E Walpole
- Princess Alexandra Hospital, Brisbane, Australia; University of Queensland, Brisbane, Australia
| | - M S Carlino
- Melanoma Institute Australia, The University of Sydney, Mater Hospital, Sydney, Australia; Westmead and Blacktown Hospitals, Melanoma Institute Australia, Sydney, Australia; University of Sydney, Sydney, Australia
| | - C Lebbe
- Université de Paris, AP-HP Dermatology and CIC, INSERM U976, Saint Louis Hospital, Paris, France
| | - M Sznol
- Yale Cancer Center, New Haven, USA
| | - E Jensen
- Merck & Co., Inc., Kenilworth, USA
| | | | | | - C Robert
- Department of Oncology, Service of Dermatology, Gustave Roussy, Villejuif, France; Paris-Saclay University, Orsay, France
| |
Collapse
|
26
|
Katsandris A, Ziogas DC, Kontouri M, Staikoglou S, Gogas H. Atezolizumab plus vemurafenib and cobimetinib for the treatment of BRAF V600-mutant advanced melanoma: from an hypothetic triplet to an approved regimen. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1976637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Aikaterini Katsandris
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Dimitrios C. Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Maria Kontouri
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Stavroula Staikoglou
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| |
Collapse
|
27
|
Jung T, Haist M, Kuske M, Grabbe S, Bros M. Immunomodulatory Properties of BRAF and MEK Inhibitors Used for Melanoma Therapy-Paradoxical ERK Activation and Beyond. Int J Mol Sci 2021; 22:ijms22189890. [PMID: 34576054 PMCID: PMC8469254 DOI: 10.3390/ijms22189890] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/21/2022] Open
Abstract
The advent of mitogen-activated protein kinase (MAPK) inhibitors that directly inhibit tumor growth and of immune checkpoint inhibitors (ICI) that boost effector T cell responses have strongly improved the treatment of metastatic melanoma. In about half of all melanoma patients, tumor growth is driven by gain-of-function mutations of BRAF (v-rat fibrosarcoma (Raf) murine sarcoma viral oncogene homolog B), which results in constitutive ERK activation. Patients with a BRAF mutation are regularly treated with a combination of BRAF and MEK (MAPK/ERK kinase) inhibitors. Next to the antiproliferative effects of BRAF/MEKi, accumulating preclinical evidence suggests that BRAF/MEKi exert immunomodulatory functions such as paradoxical ERK activation as well as additional effects in non-tumor cells. In this review, we present the current knowledge on the immunomodulatory functions of BRAF/MEKi as well as the non-intended effects of ICI and discuss the potential synergistic effects of ICI and MAPK inhibitors in melanoma treatment.
Collapse
|
28
|
Gumusay O, Wabl CA, Rugo HS. Trials of Immunotherapy in Triple Negative Breast Cancer. CURRENT BREAST CANCER REPORTS 2021. [DOI: 10.1007/s12609-021-00418-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
29
|
de Souza N, de Oliveira ÉA, Faião-Flores F, Pimenta LA, Quincoces JAP, Sampaio SC, Maria-Engler SS. Metalloproteinases Suppression Driven by the Curcumin Analog DM-1 Modulates Invasion in BRAF-Resistant Melanomas. Anticancer Agents Med Chem 2021; 20:1038-1050. [PMID: 32067622 DOI: 10.2174/1871520620666200218111422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/15/2020] [Accepted: 01/31/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Melanoma is the most aggressive skin cancer, and BRAF (V600E) is the most frequent mutation that led to the development of BRAF inhibitors (BRAFi). However, patients treated with BRAFi usually present recidivism after 6-9 months. Curcumin is a turmeric substance, and it has been deeply investigated due to its anti-inflammatory and antitumoral effects. Still, the low bioavailability and biodisponibility encouraged the investigation of different analogs. DM-1 is a curcumin analog and has shown an antitumoral impact in previous studies. METHODS Evaluated DM-1 stability and cytotoxic effects for BRAFi-sensitive and resistant melanomas, as well as the role in the metalloproteinases modulation. RESULTS DM-1 showed growth inhibitory potential for melanoma cells, demonstrated by reduction of colony formation, migration and endothelial tube formation, and cell cycle arrest. Subtoxic doses were able to downregulate important Metalloproteinases (MMPs) related to invasiveness, such as MMP-1, -2 and -9. Negative modulations of TIMP-2 and MMP-14 reduced MMP-2 and -9 activity; however, the reverse effect is seen when increased TIMP-2 and MMP-14 resulted in raised MMP-2. CONCLUSION These findings provide essential details into the functional role of DM-1 in melanomas, encouraging further studies in the development of combinatorial treatments for melanomas.
Collapse
Affiliation(s)
- Nayane de Souza
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| | - Érica Aparecida de Oliveira
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| | - Fernanda Faião-Flores
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| | | | - José A P Quincoces
- Laboratory of Organic Synthesis, Anhanguera University of São Paulo, UNIAN, Sao Paulo, Brazil
| | - Sandra C Sampaio
- Butantan Institute, Pathophysiology Laboratory, Sao Paulo, Brazil
| | - Silvya S Maria-Engler
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| |
Collapse
|
30
|
Discontinuation of BRAF/MEK-Directed Targeted Therapy after Complete Remission of Metastatic Melanoma-A Retrospective Multicenter ADOReg Study. Cancers (Basel) 2021; 13:cancers13102312. [PMID: 34065877 PMCID: PMC8151093 DOI: 10.3390/cancers13102312] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
The advent of BRAF/MEK inhibitors (BRAFi/MEKi) has significantly improved progression-free (PFS) and overall survival (OS) for patients with advanced BRAF-V600-mutant melanoma. Long-term survivors have been identified particularly among patients with a complete response (CR) to BRAF/MEK-directed targeted therapy (TT). However, it remains unclear which patients who achieved a CR maintain a durable response and whether treatment cessation might be a safe option in these patients. Therefore, this study investigated the impact of treatment cessation on the clinical course of patients with a CR upon BRAF/MEK-directed-TT. We retrospectively selected patients with BRAF-V600-mutant advanced non-resectable melanoma who had been treated with BRAFi ± MEKi therapy and achieved a CR upon treatment out of the multicentric skin cancer registry ADOReg. Data on baseline patient characteristics, duration of TT, treatment cessation, tumor progression (TP) and response to second-line treatments were collected and analyzed. Of 461 patients who received BRAF/MEK-directed TT 37 achieved a CR. TP after initial CR was observed in 22 patients (60%) mainly affecting patients who discontinued TT (n = 22/26), whereas all patients with ongoing TT (n = 11) maintained their CR. Accordingly, patients who discontinued TT had a higher risk of TP compared to patients with ongoing treatment (p < 0.001). However, our data also show that patients who received TT for more than 16 months and who discontinued TT for other reasons than TP or toxicity did not have a shorter PFS compared to patients with ongoing treatment. Response rates to second-line treatment being initiated in 21 patients, varied between 27% for immune-checkpoint inhibitors (ICI) and 60% for BRAFi/MEKi rechallenge. In summary, we identified a considerable number of patients who achieved a CR upon BRAF/MEK-directed TT in this contemporary real-world cohort of patients with BRAF-V600-mutant melanoma. Sustained PFS was not restricted to ongoing TT but was also found in patients who discontinued TT.
Collapse
|
31
|
Ziogas DC, Konstantinou F, Bouros S, Theochari M, Gogas H. Combining BRAF/MEK Inhibitors with Immunotherapy in the Treatment of Metastatic Melanoma. Am J Clin Dermatol 2021; 22:301-314. [PMID: 33765322 DOI: 10.1007/s40257-021-00593-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
The management and prognosis of BRAF-mutant metastatic melanoma have changed drastically following the introduction of immune checkpoint inhibitors and molecularly targeted agents. These treatment options present different mechanisms of action and toxicities but also totally distinct kinetics of their response, including a "relatively" short-lasting benefit in subsets of patients treated with BRAF/MEK inhibitors and a lower response rate in patients treated with immune checkpoint inhibitors. BRAF/MEK inhibitors, when administered prior to or concurrently with immune checkpoint inhibitors, at least transiently alter some immunosuppressive parameters of the tumor microenvironment and theoretically improve sensitivity to immunotherapy. Preclinical data from mouse models with oncogene-addicted melanoma confirmed this beneficial immune/targeted synergy and supported the clinical testing of combinations of BRAF/MEK inhibitors and immune checkpoint inhibitors to improve the activity of upfront anti-melanoma therapies. The first positive phase III results were published in 2020, and triggered the discussion about the benefits, the limitations, as well as the possible implications of combining or sequencing targeted therapies with immune checkpoint inhibitors in everyday practice. Beginning from the interplay of immune/targeted agents within the melanoma microenvironment, this review outlines available information from the retrospective experience up to the late-stage randomized evidence on combinatorial treatments. Many clinical trials are currently underway exploring open questions about optimal timing, new immune biomarkers, and eligible patient subsets for these immune/targeted regimens. Awaiting these results, decision making in the first-line setting for BRAF-mutant melanoma is still guided by the patients' characteristics and the biological aspects of melanoma.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Frosso Konstantinou
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Spyros Bouros
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Maria Theochari
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Helen Gogas
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece.
| |
Collapse
|
32
|
Shadbad MA, Hajiasgharzadeh K, Derakhshani A, Silvestris N, Baghbanzadeh A, Racanelli V, Baradaran B. From Melanoma Development to RNA-Modified Dendritic Cell Vaccines: Highlighting the Lessons From the Past. Front Immunol 2021; 12:623639. [PMID: 33692796 PMCID: PMC7937699 DOI: 10.3389/fimmu.2021.623639] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Although melanoma remains the deadliest skin cancer, the current treatment has not resulted in the desired outcomes. Unlike chemotherapy, immunotherapy has provided more tolerable approaches and revolutionized cancer therapy. Although dendritic cell-based vaccines have minor side effects, the undesirable response rates of traditional approaches have posed questions about their clinical translation. The immunosuppressive tumor microenvironment can be the underlying reason for their low response rates. Immune checkpoints and indoleamine 2,3-dioxygenase have been implicated in the induction of immunosuppressive tumor microenvironment. Growing evidence indicates that the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase/Protein kinase B (PKB) (PI3K/AKT) pathways, as the main oncogenic pathways of melanoma, can upregulate the tumoral immune checkpoints, like programmed death-ligand 1. This study briefly represents the main oncogenic pathways of melanoma and highlights the cross-talk between these oncogenic pathways with indoleamine 2,3-dioxygenase, tumoral immune checkpoints, and myeloid-derived suppressor cells. Moreover, this study sheds light on a novel tumor antigen on melanoma, which has substantial roles in tumoral immune checkpoints expression, indoleamine 2,3-dioxygenase secretion, and stimulating the oncogenic pathways. Finally, this review collects the lessons from the previous unsuccessful trials and integrates their lessons with new approaches in RNA-modified dendritic cell vaccines. Unlike traditional approaches, the advances in single-cell RNA-sequencing techniques and RNA-modified dendritic cell vaccines along with combined therapy of the immune checkpoint inhibitors, indoleamine 2,3-dioxygenase inhibitor, and RNA-modified dendritic cell-based vaccine can overcome these auto-inductive loops and pave the way for developing robust dendritic cell-based vaccines with the most favorable response rate and the least side effects.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/adverse effects
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/therapeutic use
- Cancer Vaccines/adverse effects
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Humans
- Immune Checkpoint Proteins/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/metabolism
- Melanoma/therapy
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- RNA, Small Interfering/adverse effects
- RNA, Small Interfering/genetics
- RNA, Small Interfering/immunology
- RNA, Small Interfering/therapeutic use
- Signal Transduction
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/therapy
- Tumor Escape
- Tumor Microenvironment
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/therapeutic use
- mRNA Vaccines
Collapse
Affiliation(s)
- Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
| | - Nicola Silvestris
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Wang M, Wang S, Desai J, Trapani JA, Neeson PJ. Therapeutic strategies to remodel immunologically cold tumors. Clin Transl Immunology 2020; 9:e1226. [PMID: 35136604 PMCID: PMC8809427 DOI: 10.1002/cti2.1226] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) induce a durable response in a wide range of tumor types, but only a minority of patients outside these 'responsive' tumor types respond, with some totally resistant. The primary predictor of intrinsic immune resistance to ICIs is the complete or near-complete absence of lymphocytes from the tumor, so-called immunologically cold tumors. Here, we propose two broad approaches to convert 'cold' tumors into 'hot' tumors. The first is to induce immunogenic tumor cell death, through the use of oncolytic viruses or bacteria, conventional cancer therapies (e.g. chemotherapy or radiation therapy) or small molecule drugs. The second approach is to target the tumor microenvironment, and covers diverse options such as depleting immune suppressive cells; inhibiting transforming growth factor-beta; remodelling the tumor vasculature or hypoxic environment; strengthening the infiltration and activation of antigen-presenting cells and/or effector T cells in the tumor microenvironment with immune modulators; and enhancing immunogenicity through personalised cancer vaccines. Strategies that successfully modify cold tumors to overcome their resistance to ICIs represent mechanistically driven approaches that will ultimately result in rational combination therapies to extend the clinical benefits of immunotherapy to a broader cancer cohort.
Collapse
Affiliation(s)
- Minyu Wang
- Cancer Immunology ProgramPeter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVICAustralia
- Centre for Cancer ImmunotherapyPeter Mac and VCCC allianceMelbourneVICAustralia
| | - Sen Wang
- South Australian Genomics CentreSouth Australian Health and Medical Research InstituteAdelaideSAAustralia
- Medical Genomics PlatformHudson Institute of Medical ResearchClaytonVICAustralia
| | - Jayesh Desai
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVICAustralia
- Division of Medical OncologyPeter MacCallum Cancer CentreMelbourneVICAustralia
| | - Joseph A Trapani
- Cancer Immunology ProgramPeter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVICAustralia
- Centre for Cancer ImmunotherapyPeter Mac and VCCC allianceMelbourneVICAustralia
| | - Paul J Neeson
- Cancer Immunology ProgramPeter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVICAustralia
- Centre for Cancer ImmunotherapyPeter Mac and VCCC allianceMelbourneVICAustralia
| |
Collapse
|
34
|
Dummer R, Ascierto PA, Nathan P, Robert C, Schadendorf D. Rationale for Immune Checkpoint Inhibitors Plus Targeted Therapy in Metastatic Melanoma: A Review. JAMA Oncol 2020; 6:1957-1966. [PMID: 32970096 DOI: 10.1001/jamaoncol.2020.4401] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance In recent years, the management of metastatic melanoma has been transformed by the emergence of immune checkpoint inhibitors and targeted therapies that significantly improve patient survival. The complementary response kinetics of these treatment approaches, supported by mechanistic evidence that targeted therapy affects immune aspects of the tumor microenvironment, suggest that the optimal combination or sequencing of immune checkpoint inhibitors and targeted therapy may provide additional clinical benefit. Observations Clinical responses to BRAF and/or MEK inhibitors are associated with immune changes within the tumor microenvironment that have the potential to increase the sensitivity of BRAF V600-mutant melanoma to immune checkpoint inhibitors. The combination of immune checkpoint inhibitors with targeted therapy may therefore increase duration of response, improve tumor control, extend survival, and increase the proportion of patients experiencing durable benefit. A targeted therapy-immune checkpoint inhibitor sequencing approach may also be supported by this evidence, but clinical questions regarding optimal timing, duration, and patient selection remain. Conclusions and Relevance This review outlines the rationale and preclinical evidence that support immune checkpoint inhibitor plus targeted therapy combination and sequencing strategies in melanoma and highlights the results available to date from clinical trials exploring these approaches to treatment. Several late-stage trials are under way looking to answer open questions in this field and address the continuing debate surrounding up-front combination vs sequencing. As phase 3 data have begun to emerge, trial designs and available data from key studies are discussed in the context of their resultant implications for clinical practice.
Collapse
Affiliation(s)
- Reinhard Dummer
- University Hospital Zürich Skin Cancer Center, Zürich, Switzerland
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale," Naples, Italy
| | - Paul Nathan
- Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - Caroline Robert
- Gustave Roussy and Paris-Sud-Paris-Saclay University, Villejuif, France
| | - Dirk Schadendorf
- University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
35
|
Evrard D, Hourseau M, Couvelard A, Paradis V, Gauthier H, Raymond E, Halimi C, Barry B, Faivre S. PD-L1 expression in the microenvironment and the response to checkpoint inhibitors in head and neck squamous cell carcinoma. Oncoimmunology 2020; 9:1844403. [PMID: 33299655 PMCID: PMC7714503 DOI: 10.1080/2162402x.2020.1844403] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In head and neck squamous cell carcinoma (HNSCC), data from studies using checkpoint-inhibiting antibodies that target programmed death 1 (PD-1) or its ligand the programmed death ligand 1 (PD-L1) demonstrated outstanding clinical activity. Translational investigations also suggested some correlations between therapeutic response and PD-L1 expression in tumor tissue. We comprehensively summarize results that have evaluated PD-L1 expression in HNSCC. We discuss flaws and strength of current PD-1/PD-L1 detection, quantification methods and the evaluation of PD-L1 as a prognostic and theragnostic biomarker. Understanding tumor microenvironment may help understanding resistance to checkpoint inhibitors, designing clinical trials that can exploit drug combinations.
Collapse
Affiliation(s)
- D Evrard
- Department of Otorhinolaryngology, Bichat Hospital, Paris, France
| | - M Hourseau
- Pathology Department, Bichat Hospital, Paris 7 University, Paris, France
| | - A Couvelard
- Pathology Department, Bichat Hospital, Paris 7 University, Paris, France
| | - V Paradis
- Pathology Department, Beaujon Hospital, Paris 7 University, Paris, France
| | - H Gauthier
- Medical Oncology Department, Saint-Louis Hospital, Paris 7 University, Paris, France
| | - E Raymond
- Medical Oncology Department, Paris-St Joseph Hospital, Paris, France
| | - C Halimi
- Department of Otorhinolaryngology, Bichat Hospital, Paris, France
| | - B Barry
- Department of Otorhinolaryngology, Bichat Hospital, Paris, France
| | - S Faivre
- Medical Oncology Department, Saint-Louis Hospital, Paris 7 University, Paris, France
| |
Collapse
|
36
|
Combined PD-1, BRAF and MEK inhibition in advanced BRAF-mutant melanoma: safety run-in and biomarker cohorts of COMBI-i. Nat Med 2020; 26:1557-1563. [PMID: 33020648 DOI: 10.1038/s41591-020-1082-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022]
Abstract
Immune and targeted therapies achieve long-term survival in metastatic melanoma; however, new treatment strategies are needed to improve patients' outcomes1,2. We report on the efficacy, safety and biomarker analysis from the single-arm safety run-in (part 1; n = 9) and biomarker (part 2; n = 27) cohorts of the randomized, placebo-controlled, phase 3 COMBI-i trial (NCT02967692) of the anti-PD-1 antibody spartalizumab, in combination with the BRAF inhibitor dabrafenib and MEK inhibitor trametinib. Patients (n = 36) had previously untreated BRAF V600-mutant unresectable or metastatic melanoma. In part 1, the recommended phase 3 regimen was identified based on the incidence of dose-limiting toxicities (DLTs; primary endpoint): 400 mg of spartalizumab every 4 weeks plus 150 mg of dabrafenib twice daily plus 2 mg of trametinib once daily. Part 2 characterized changes in PD-L1 levels and CD8+ cells following treatment (primary endpoint), and analyzed additional biomarkers. Assessments of efficacy and safety were key secondary endpoints (median follow-up, 24.3 months). Spartalizumab plus dabrafenib and trametinib led to an objective response rate (ORR) of 78%, including 44% complete responses (CRs). Grade ≥3 treatment-related adverse events (TRAEs) were experienced by 72% of patients. All patients had temporary dose modifications, and 17% permanently discontinued all three study drugs due to TRAEs. Early progression-free survival (PFS) events were associated with low tumor mutational burden/T cell-inflamed gene expression signature (GES) or high immunosuppressive tumor microenvironment (TME) GES levels at baseline; an immunosuppressive TME may also preclude CR. Overall, the efficacy, safety and on-treatment biomarker modulations associated with spartalizumab plus dabrafenib and trametinib are promising, and biomarkers that may predict long-term benefit were identified.
Collapse
|
37
|
Bai X, Flaherty KT. Targeted and immunotherapies in BRAF mutant melanoma: where we stand and what to expect. Br J Dermatol 2020; 185:253-262. [PMID: 32652567 DOI: 10.1111/bjd.19394] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2020] [Indexed: 01/01/2023]
Abstract
The therapeutic landscape for melanoma has evolved drastically in the past decade. Currently, immune checkpoint inhibitors and small-molecule inhibitors targeting the mitogen-activated protein kinase (MAPK) pathway are the two mainstay therapies for BRAFV600 mutant advanced melanoma. Although MAPK dependence has been variably demonstrated in melanomas lacking BRAFV600 mutations, definitive evidence of benefit with MAPK inhibitors has not been demonstrated. Thus, in the BRAFV600 'wild-type' setting, immune checkpoint inhibitors are the standalone option(s). In the BRAFV600 mutant setting, there is no definitive evidence prioritizing one therapeutic modality over another. Herein, we review the updated data of the pivotal phase III randomized controlled trials that established the standard-of-care first-line treatment for advanced melanoma, as it provides insights into long-term benefit, which is a major factor in therapy selection. We discuss the clinical considerations for choosing between these therapies in the front-line setting and beyond, specifically for patients with BRAFV600 mutant melanoma based on currently available evidence. We have previously proposed a time-dependent resistance paradigm in which future therapeutic development strategies can be rooted. We also discuss how these Food and Drug Administration (FDA)-approved therapeutic modalities are being pursued earlier in the course of disease management, namely in adjuvant and neoadjuvant settings. FDA-approved interlesional oncolytic virotherapy in the modern era is also briefly discussed.
Collapse
Affiliation(s)
- X Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China.,Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - K T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Jiang P, He S, Li Y, Xu Z. Identification of Therapeutic and Prognostic Biomarkers of Lamin C (LAMC) Family Members in Head and Neck Squamous Cell Carcinoma. Med Sci Monit 2020; 26:e925735. [PMID: 32860673 PMCID: PMC7477928 DOI: 10.12659/msm.925735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSC) is an invasive malignancy with a high worldwide mortality, despite considerable recent advancements in diagnosis and treatment. Increasing evidence indicates that the Lamin C (LAMC) gene family is associated with the progression of diverse cancers, nevertheless, this association is not well understood. Material/Methods A systematic study addressing the expression and prognostic value of LAMC, and the relationship between LAMC and tumor immune response in HNSC was done. Finally, we performed drug screening to identify specific drugs. Results Compared to normal samples, expressions of LAMC1 and LAMC2 were significantly increased in HNSC, and LAMC2 was obviously correlated with an adverse prognosis for patients. LAMC2 expression level was significantly correlated with the infiltrating levels of B cells, CD8+ T cells, CD4+ T cells, and macrophages. Moreover, LAMC2 exhibited strong correlations with diverse immune markers, immune microenvironment, and immune checkpoint molecules. Finally, candidate drugs that targeted LAMC2 were identified. Conclusions This study suggests that LAMC2 could serve as a new prognostic biomarker, and it could be used for efficacy of target for immune response and for drug sensitivity prediction in HNSC.
Collapse
Affiliation(s)
- Pan Jiang
- Department of Stomatology, The Third People Hospital of Hainan Province, Sanya, Hainan, China (mainland)
| | - Shengteng He
- Department of Stomatology, The Third People Hospital of Hainan Province, Sanya, Hainan, China (mainland)
| | - Yanli Li
- Department of Stomatology, The Third People Hospital of Hainan Province, Sanya, Hainan, China (mainland)
| | - Zheng Xu
- Department of Stomatology, The Third People Hospital of Hainan Province, Sanya, Hainan, China (mainland)
| |
Collapse
|
39
|
Tanda ET, Vanni I, Boutros A, Andreotti V, Bruno W, Ghiorzo P, Spagnolo F. Current State of Target Treatment in BRAF Mutated Melanoma. Front Mol Biosci 2020; 7:154. [PMID: 32760738 PMCID: PMC7371970 DOI: 10.3389/fmolb.2020.00154] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/19/2020] [Indexed: 12/17/2022] Open
Abstract
Incidence of melanoma has been constantly growing during the last decades. Although most of the new diagnoses are represented by thin melanomas, the number of melanoma-related deaths in 2018 was 60,712 worldwide (Global Cancer Observatory, 2019). Until 2011, no systemic therapy showed to improve survival in patients with advanced or metastatic melanoma. At that time, standard of care was chemotherapy, with very limited results. The identification of BRAF V600 mutation, and the subsequent introduction of BRAF targeting drugs, radically changed the clinical practice and dramatically improved outcomes. In this review, we will retrace the development of molecular-target drugs and the current therapeutic scenario for patients with BRAF mutated melanoma, from the introduction of BRAF inhibitors as single agents to modern clinical practice. We will also discuss the resistance mechanisms identified so far, and the future therapeutic perspectives in BRAF mutated melanoma.
Collapse
Affiliation(s)
| | - Irene Vanni
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | - Andrea Boutros
- Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Virginia Andreotti
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | - William Bruno
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | - Paola Ghiorzo
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | | |
Collapse
|
40
|
Isazadeh A, Hajazimian S, Garshasbi H, Shadman B, Baghbanzadeh A, Chavoshi R, Taefehshokr S, Farhoudi Sefidan Jadid M, Hajiasgharzadeh K, Baradaran B. Resistance mechanisms to immune checkpoints blockade by monoclonal antibody drugs in cancer immunotherapy: Focus on myeloma. J Cell Physiol 2020; 236:791-805. [PMID: 32592235 DOI: 10.1002/jcp.29905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022]
Abstract
Multiple myeloma (MM) is a clonal B-cell malignancy characterized by the accumulation of neoplastic proliferation of a plasma cell in the bone marrow that produces a monoclonal immunoglobulin. The immune checkpoint inhibitors against programmed death-1/programmed death-1 ligand and cytotoxic T-lymphocyte antigen 4 axis have demonstrated appropriate anticancer activity in several solid tumors and liquid cancers, and are rapidly transforming the practice of medical oncology. However, in a high percentage of patients, the efficacy of immune checkpoints blockade remains limited due to innate or primary resistance. Moreover, the malignancies progress in many patients due to acquired or secondary resistance, even after the clinical response to immune checkpoints' blockade. The evidence shows that multiple tumor-intrinsic and tumor-extrinsic factors and alterations in signaling pathways are involved in primary and secondary resistance to immune checkpoints blockade. Improved identification of intrinsic and extrinsic factors and mechanisms of resistance or response to immune checkpoints blockade may not only provide novel prognostic or predictive biomarkers but also guide the optimal combination/sequencing of immune checkpoint blockade therapy in the clinic. Here, we review the underlying biology and role of immune checkpoints blockade in patients with MM. Furthermore, we review the host and tumor-related factor effects on immune checkpoints blockade in MM immunotherapy.
Collapse
Affiliation(s)
- Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Hajazimian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Garshasbi
- Department of Genetic, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Behrouz Shadman
- Ege University Medical School, Department of Medical Biology, Izmir, Turkey
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Chavoshi
- Department of Genetic, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Xu J, Wang F, Yan Y, Zhang Y, Du Y, Sun G. Prognostic and Clinicopathological Value of PD-L1 in Melanoma: A Meta-Analysis. Am J Med Sci 2020; 359:339-346. [PMID: 32498941 DOI: 10.1016/j.amjms.2020.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/08/2020] [Accepted: 03/25/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND There is a growing interest in using programmed death ligand-1 (PD-L1) as a prognostic marker for melanoma. We conducted this meta-analysis to explore the prognostic and clinicopathological value of PD-L1 in melanoma. MATERIALS AND METHODS The electronic databases PubMed, Web of Science and the Cochrane Library were searched for relevant studies. The major investigated parameters were PD-L1 expression levels in relation to patient gender, tumor-infiltrating lymphocytes (TILs), tumor stage, lymph node (LN) metastasis, histological type, progression-free survival (PFS) and overall survival (OS). Odds ratios (ORs) and hazard ratios (HRs) were computed using the fixed-effect or random-effects model according to data heterogeneity. RESULTS Positive PD-L1 expression was significantly associated with high levels of TILs (OR = 7.56, 95% CI 2.04-28.02), metastatic melanoma (OR = 0.45, 95% CI 0.30-0.67) and LN-positive melanoma (OR = 2.56, 95% CI 1.31-4.99) but not gender or histological type. In addition, the pooled HRs showed no relation between PD-L1 expression and PFS (HR = 1.18, 95% CI 0.83-1.69) or OS (HR = 0.77, 95% CI 0.47-1.25). When restricted to metastatic melanoma, positive PD-L1 expression was significantly related to prolonged OS (HR = 0.57, 95% CI 0.46-0.70). CONCLUSIONS Positive PD-L1 expression may be an important prognostic factor for longer OS in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Jing Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fang Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunfang Yan
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yiruo Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yingying Du
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
42
|
Nukui A, Kamai T, Arai K, Kijima T, Kobayashi M, Narimatsu T, Kambara T, Yuki H, Betsunoh H, Abe H, Fukabori Y, Yashi M, Yoshida KI. Association of cancer progression with elevated expression of programmed cell death protein 1 ligand 1 by upper tract urothelial carcinoma and increased tumor-infiltrating lymphocyte density. Cancer Immunol Immunother 2020; 69:689-702. [PMID: 32030476 PMCID: PMC7183489 DOI: 10.1007/s00262-020-02499-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
Background Increased expression of programmed cell death 1 ligand 1 (PD-L1) by tumor cells is thought to be a mechanism through which solid cancers promote immune tolerance. However, the association between PD-L1 expression and the prognosis of upper urinary tract urothelial carcinoma (UTUC) remains unknown. Methods We examined immunohistochemical PD-L1 expression and the tumor-infiltrating lymphocyte density (TILD) in 79 patients with UTUC who underwent nephroureterectomy. We classified the tumors into four types based on the combination of PD-L1 expression and TILD, and studied the clinicopathological characteristics of these four tumor types. Results Elevated expression of PD-L1 by tumor cells and a higher TILD were associated with a worse histological grade, higher pT stage, and higher peripheral blood neutrophil-to-lymphocyte ratio. Elevated expression of PD-L1 by tumor cells, a higher TILD, and type I, III, or IV tumors with elevated expression of either PD-L1 or TILD showed a positive correlation with poorer differentiation and local invasion. These three variables were associated with shorter progression-free survival and overall survival in univariate analysis, but only the latter was an independent determinant according to multivariate analysis. The patients who had type II tumors with lower PD-L1 expression and a lower TILD showed more favorable survival than the other three groups. Conclusions These findings suggest that PD-L1 expression and TILs in the tumor microenvironment influence the progression of UTUC. Accordingly, it is important to understand the immunologic characteristics of the tumor microenvironment to develop more effective treatment strategies for this cancer. Electronic supplementary material The online version of this article (10.1007/s00262-020-02499-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Akinori Nukui
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Takao Kamai
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan.
| | - Kyoko Arai
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Toshiki Kijima
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Minoru Kobayashi
- Department of Urology, Utsunomiya Memorial Hospital, Utsunomiya, Tochigi, Japan
| | - Takahiro Narimatsu
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Tsunehito Kambara
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Hideo Yuki
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Hironori Betsunoh
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Hideyuki Abe
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Yoshitatsu Fukabori
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Masahiro Yashi
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| | - Ken-Ichiro Yoshida
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi Mibu, Utsunomiya, Tochigi, 321-0293, Japan
| |
Collapse
|
43
|
Ziogas DC, Konstantinou F, Bouros S, Gogas H. Identifying the optimum first-line therapy in BRAF-mutant metastatic melanoma. Expert Rev Anticancer Ther 2020; 20:53-62. [PMID: 31903803 DOI: 10.1080/14737140.2020.1711737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: The emergence of molecularly targeted agents and immune checkpoint inhibitors has positively revolutionized the management and prognosis of BRAF-mutant metastatic melanoma. However, the availability of both therapeutic options, with their pros and cons, rationally triggered clinical considerations for the optimum frontline and subsequent treatment decisions.Areas covered: Here, we debate all approved therapies in patients with BRAF-mutant metastatic melanoma evaluating their efficacy and safety based on their pivotal trials. With prospective randomized data pending, retrospective comparisons of BRAF/MEK versus immune checkpoint inhibitors are reviewed to recognize any advantage between these two alternatives and to optimize their implementation. Preclinical and early clinical results of combining concurrently or sequentially targeted therapy and immunotherapy are also discussed.Expert opinion: BRAF/MEK inhibitors produce rapid and deep responses and should be included in first-line approaches, particularly in cases with aggressive and bulky disease, while single or double checkpoint inhibition lead to more durable responses and could be involved either in frontline treatment of BRAF-mutant melanoma with less unfavorable characteristics or in maintenance after initial targeted induction or in future immune/targeted regimens for high-risk groups. Data from ongoing trials directly comparing or combining these strategies are expected to update their role in a more individualized basis.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Frosso Konstantinou
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Spyros Bouros
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| |
Collapse
|
44
|
Martin AM, Bell WR, Yuan M, Harris L, Poore B, Arnold A, Engle EL, Asnaghi L, Lim M, Raabe EH, Eberhart CG. PD-L1 Expression in Pediatric Low-Grade Gliomas Is Independent of BRAF V600E Mutational Status. J Neuropathol Exp Neurol 2020; 79:74-85. [PMID: 31819973 PMCID: PMC8660581 DOI: 10.1093/jnen/nlz119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/04/2019] [Accepted: 11/01/2019] [Indexed: 01/01/2023] Open
Abstract
To evaluate a potential relationship between BRAF V600E mutation and PD-L1 expression, we examined the expression of PD-L1 in pediatric high- and low-grade glioma cell lines as well as a cohort of pediatric low-grade glioma patient samples. Half of the tumors in our patient cohort were V600-wildtype and half were V600E mutant. All tumors expressed PD-L1. In most tumors, PD-L1 expression was low (<5%), but in some cases over 50% of cells were positive. Extent of PD-L1 expression and immune cell infiltration was independent of BRAF V600E mutational status. All cell lines evaluated, including a BRAF V600E mutant xenograft, expressed PD-L1. Transient transfection of cell lines with a plasmid expressing mutant BRAF V600E had minimal effect on PD-L1 expression. These findings suggest that the PD-1 pathway is active in subsets of pediatric low-grade glioma as a mechanism of immune evasion independent of BRAF V600E mutational status. Low-grade gliomas that are unresectable and refractory to traditional therapy are associated with significant morbidity and continue to pose a treatment challenge. PD-1 pathway inhibitors may offer an alternative treatment approach. Clinical trials will be critical in determining whether PD-L1 expression indicates likely therapeutic benefit with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Allison M Martin
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - W Robert Bell
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ming Yuan
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lauren Harris
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Bradley Poore
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Antje Arnold
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Elizabeth L Engle
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Laura Asnaghi
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michael Lim
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Eric H Raabe
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Charles G Eberhart
- Division of Pediatric Oncology, Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Baltimore, Maryland (AMM, EHR); Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota (WRB); Department of Pathology, Division of Neuropathology, Johns Hopkins School of Medicine, Baltimore, Maryland (MY, BP, AA, LA, EHR, CGE); Department of Molecular and Cell Biology, The Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland (LH); Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy (ELE); and Department of Neurosurgery, Division of Neurosurgical Oncology (ML), Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
45
|
Carr MJ, Sun J, Eroglu Z, Zager JS. An evaluation of encorafenib for the treatment of melanoma. Expert Opin Pharmacother 2019; 21:155-161. [PMID: 31790307 DOI: 10.1080/14656566.2019.1694664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: In the treatment of advanced BRAF-mutant melanoma, selective regulation of the MAPK pathway with BRAF and MEK inhibition has emerged as one of the mainstays of therapy.Areas covered: The authors present the current data on encorafenib as a compound, its pharmacokinetic and pharmacodynamics properties. This review includes current data on encorafenib therapy as a single agent as well as in combination with the MEK-inhibitor binimetinib and other systemic therapies.Expert opinion: BRAF inhibition with encorafenib exhibits substantial antitumor activity with less paradoxical MAPK pathway activation leading to treatment resistance. Combination therapy with MEK inhibitors improves response rate, progression-free survival, and overall survival in patients with BRAF-mutant metastatic melanoma compared to prior treatment regimens. Serious adverse events, including the development of cutaneous malignancies, are reported at lower rates with combination therapy, while less severe events such as pyrexia can be more common. Existing data is lacking for a recommendation of triplet therapy, although results from multiple ongoing trials are highly anticipated.
Collapse
Affiliation(s)
- Michael J Carr
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - James Sun
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
46
|
Han RH, Dunn GP, Chheda MG, Kim AH. The impact of systemic precision medicine and immunotherapy treatments on brain metastases. Oncotarget 2019; 10:6739-6753. [PMID: 31803366 PMCID: PMC6877099 DOI: 10.18632/oncotarget.27328] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
Metastases from melanoma, lung and breast cancer are among the most common causes of intracranial malignancy. Standard of care for brain metastases include a combination of surgical resection, stereotactic radiosurgery, and whole-brain radiation. However, evidence continues to accumulate regarding the efficacy of molecularly-targeted systemic treatments and immunotherapy. For non-small cell lung cancer (NSCLC), numerous clinical trials have demonstrated intracranial activity for inhibitors of EGFR and ALK. Patients with melanoma brain metastases may benefit from systemic therapy using BRAF-inhibitors with and without trametinib. Several targeted options are available for breast cancer brain metastases that overexpress HER2, although agents with intracranial activity are still needed for other molecular subtypes. Immune checkpoint inhibitors including anti-CTLA-4 and anti-PD-1/PD-L1 antibodies are yielding impressive responses in intracranial manifestations of metastatic melanoma and NSCLC. Given the promising early results with these emerging therapies, management of eligible patients will require increased multidisciplinary discussion incorporating novel systemic treatment approaches prior or in addition to local therapy.
Collapse
Affiliation(s)
- Rowland H Han
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Milan G Chheda
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
47
|
Mason R, Dearden HC, Nguyen B, Soon JA, Smith JL, Randhawa M, Mant A, Warburton L, Lo S, Meniawy T, Guminski A, Parente P, Ali S, Haydon A, Long GV, Carlino MS, Millward M, Atkinson VG, Menzies AM. Combined ipilimumab and nivolumab first-line and after BRAF-targeted therapy in advanced melanoma. Pigment Cell Melanoma Res 2019; 33:358-365. [PMID: 31587511 DOI: 10.1111/pcmr.12831] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 09/03/2019] [Accepted: 09/12/2019] [Indexed: 11/28/2022]
Abstract
The combination of ipilimumab and nivolumab is a highly active systemic therapy for metastatic melanoma but can cause significant toxicity. We explore the safety and efficacy of this treatment in routine clinical practice, particularly in the setting of serine/threonine-protein kinase B-Raf (BRAF)-targeted therapy. Consecutive patients with unresectable stage IIIC/IV melanoma commenced on ipilimumab and nivolumab across 10 tertiary melanoma institutions in Australia were identified retrospectively. Data collected included demographics, response and survival outcomes. A total of 152 patients were included for analysis, 39% were treatment-naïve and 22% failed first-line BRAF/MEK inhibitors. Treatment-related adverse events occurred in 67% of patients, grade 3-5 in 38%. The overall objective response rate was 41%, 57% in treatment-naïve and 21% in BRAF/MEK failure patients. Median progression-free survival was 4.0 months (95% CI, 3.0-6.0) in the whole cohort, 11.0 months (95% CI, 6.0-NR) in treatment-naïve and 2.0 months (95% CI, 1.4-4.6) in BRAF/MEK failure patients. The combination of ipilimumab and nivolumab can be used safely and effectively in a real-world population. While first-line efficacy appears comparable to trial populations, BRAF-mutant patients failing prior BRAF/MEK inhibitors show less response.
Collapse
Affiliation(s)
- Robert Mason
- Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - Helen C Dearden
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Bella Nguyen
- Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Jennifer A Soon
- Alfred Hospital, Monash University, Melbourne, Vic., Australia
| | | | | | | | | | - Serigne Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Tarek Meniawy
- Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,St John of God Hospital, Subiaco, WA, Australia.,University of Western Australia, Nedlands, WA, Australia
| | - Alexander Guminski
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Royal North Shore Hospital and Mater Hospitals, Sydney, NSW, Australia
| | - Phillip Parente
- Eastern Health, Box Hill, Vic., Australia.,Monash University, Melbourne, Vic., Australia
| | - Sayed Ali
- The Canberra Hospital, Canberra, ACT, Australia.,University of Western Australia, Nedlands, WA, Australia
| | - Andrew Haydon
- Alfred Hospital, Monash University, Melbourne, Vic., Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Royal North Shore Hospital and Mater Hospitals, Sydney, NSW, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - Michael Millward
- Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,University of Western Australia, Nedlands, WA, Australia
| | - Victoria G Atkinson
- Princess Alexandra Hospital, Brisbane, Qld, Australia.,Greenslopes Private Hospital, Brisbane, Qld, Australia.,University of Queensland, Brisbane, Qld, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Royal North Shore Hospital and Mater Hospitals, Sydney, NSW, Australia
| |
Collapse
|
48
|
Zidlik V, Bezdekova M, Brychtova S. Tumor infiltrating lymphocytes in malignant melanoma - allies or foes? Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2019; 164:43-48. [PMID: 31649385 DOI: 10.5507/bp.2019.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/11/2019] [Indexed: 01/22/2023] Open
Abstract
This is an overview of current problematics regarding the role of tumor infiltrating lymphocytes (TILs) in malignant melanomas. Various and often conflicting data have been published, correlating tumor type, stage, prognosis, as well as sex and age of patients. This is partly due to heterogeneity in scaling systems and unstandardized TILs grading but also due to changes of tumor-host interactions. Melanomas are an immunologically heterogeneous group with variability of TILs, where distinct gene expression patterns were found in tumors with absent, and/or non- brisk TIL grade versus brisk TIL grade. However, the presence of TILs alone appears to be inadequate for implicating them as immunologically functional. Further characterisation of TIL phenotype and function is warranted. This especially concerns, evaluation of TILs of the suppressor phenotype but rather than as a prognostic factor, more for prediction of targeted immunotherapy.
Collapse
Affiliation(s)
- Vladimir Zidlik
- Department of Pathology, University Hospital Ostrava, Czech Republic.,Department of Pathology, CGB Laboratory, Ostrava, Czech Republic
| | - Michala Bezdekova
- Institute of Clinical and Molecular Pathology, University Hospital Olomouc, Olomouc, Czech Republic
| | - Svetlana Brychtova
- Institute of Clinical and Molecular Pathology, University Hospital Olomouc, Olomouc, Czech Republic.,Institute of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| |
Collapse
|
49
|
Fenton SE, Sosman JA, Chandra S. Resistance mechanisms in melanoma to immuneoncologic therapy with checkpoint inhibitors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:744-761. [PMID: 35582566 PMCID: PMC8992532 DOI: 10.20517/cdr.2019.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 11/14/2022]
Abstract
Checkpoint inhibitors act by blocking physiologic mechanisms coopted by tumor cells to evade immune surveillance, restoring the immune system's ability to identify and kill malignant cells. These therapies have dramatically improved outcomes in multiple tumor types with durable responses in many patients, leading to FDA approval first in advanced melanoma, then in many other malignancies. However, as experience with checkpoint inhibitors has grown, populations of patients who are primary nonresponders or develop secondary resistance have been the majority of cases, even in melanoma. Mechanisms of resistance include those inherent to the tumor microenvironment, the tumor cells themselves, and the function of the patient's native immune cells. This review will discuss resistance to checkpoint inhibitors in melanoma as well as possible methods to restore sensitivity.
Collapse
Affiliation(s)
- Sarah E. Fenton
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| | - Jeffrey A. Sosman
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| | - Sunandana Chandra
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
50
|
Rossi A, Roberto M, Panebianco M, Botticelli A, Mazzuca F, Marchetti P. Drug resistance of BRAF-mutant melanoma: Review of up-to-date mechanisms of action and promising targeted agents. Eur J Pharmacol 2019; 862:172621. [PMID: 31446019 DOI: 10.1016/j.ejphar.2019.172621] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 12/29/2022]
Abstract
Melanoma onset and progression are associated with a high variety of activating mutations in the MAPK-pathway, most frequently involving BRAF (35-45%) and NRAS (15-25%) genes, but also c-KIT and PTEN. Targeted therapies with BRAF and MEK inhibitors showed promising results over the past years, but it is known that most responses are temporary, and almost all of patients develop a tumor relapse within one year. Different drug-resistance mechanisms underlie the progression of disease and activation of both MAPK and PI3K/AKT/mTOR pathways. Therefore, in this article we reviewed the main studies about clinical effects of several target inhibitors, describing properly the most prominent mechanisms of both intrinsic and acquired resistance. Furthermore, suggestive strategies for overcoming drug resistance and the most recent alternative combination therapies to optimize the use of MAPK pathway inhibitors were also discussed.
Collapse
Affiliation(s)
- Alessandro Rossi
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Michela Roberto
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy; Department of Medical-Surgical Sciences and Translation Medicine, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy.
| | - Martina Panebianco
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy; Oncology Unit, IDI-IRCCS of Rome, Italy
| |
Collapse
|