51
|
Yin L, Liu X, Wu J, Yang J, Wang J, Dou H, Hou Y. LS-007 inhibits melanoma growth via inducing apoptosis and cell cycle arrest and regulating macrophage polarization. Melanoma Res 2022; 32:419-427. [PMID: 36094494 DOI: 10.1097/cmr.0000000000000853] [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
LS-007, an inhibitor of cyclin-dependent kinase 9 (CDK9), exhibits potential antitumor activity against chronic lymphocytic leukemia and ovarian cancer, but its effect on melanoma and tumor microenvironment (TME) has not been reported yet. This study aimed to investigate the role of LS-007 in B16F10 melanoma and relevant mechanisms. LS-007 significantly inhibited viability and induced apoptosis of B16F10 cells in a dose-dependent manner, which were accompanied with the increased ratio of Bax to Bcl-2 and decreased Mcl-1 mRNA level. Western blot analysis showed that LS-007 increased the expression of cleaved caspase-3 and poly ADP-ribose polymerase (PARP). Furthermore, flow cytometry analysis and qRT-PCR results showed that LS-007 treatment resulted in cell cycle arrest by changing cell cycle-related gene expression. Notably, in vivo evaluation showed that LS-007 significantly decreased the weight and volume of tumor and the expression of Ki67, promoted the expression of iNOS and inhibited the expression of CD206, suggesting that LS-007 might inhibit tumor growth by suppressing polarization of macrophages into tumor-associated macrophages (TAMs) in the TME. The increase in M1/M2 treated with LS-007 detected by flow cytometry hinted that macrophages were polarized towards an antitumor phenotype. In addition, LS-007 induced higher apoptotic rate of B16F10 cells when co-cultured B16F10 with BMDMs. LS-007 has inhibitory effects on B16F10 cells in vivo and in vitro via inducing apoptosis, cell cycle arrest, and changing macrophage function in the TME.
Collapse
Affiliation(s)
- Lijie Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Xinghan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Jinjin Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Jingjing Yang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Jiali Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
52
|
Fomchenko EI, Bayley JC, Alvarez-Breckenridge C, Rhines LD, Tatsui CE. Spinal Metastases and the Evolving Role of Molecular Targeted Therapy, Chemotherapy, and Immunotherapy. Neurospine 2022; 19:978-993. [PMID: 36597635 PMCID: PMC9816609 DOI: 10.14245/ns.2244290.145] [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: 04/07/2022] [Accepted: 07/13/2022] [Indexed: 12/27/2022] Open
Abstract
Metastatic involvement of the spine is a common complication of systemic cancer progression. Surgery and external beam radiotherapy are palliative treatment modalities aiming to preserve neurological function, control pain and maintain functional status. More recently, with development of image guidance and stereotactic delivery of high doses of conformal radiation, local tumor control has improved; however recurrent or radiation refractory disease remains a significant clinical problem with limited treatment options. This manuscript represents a narrative overview of novel targeted molecular therapies, chemotherapies, and immunotherapy treatments for patients with breast, lung, melanoma, renal cell, prostate, and thyroid cancers, which resulted in improved responses compared to standard chemotherapy. We present clinical examples of excellent responses in spinal metastatic disease which have not been specifically documented in the literature, as most clinical trials evaluate treatment response based on visceral disease. This review is useful for the spine surgeons treating patients with metastatic disease as knowledge of these responses could help with timing and planning of surgical interventions, as well as promote multidisciplinary discussions, allowing development of an individualized treatment strategy to patients presenting with widespread multifocal progressive disease, where surgery could lead to suboptimal results.
Collapse
Affiliation(s)
| | - James C. Bayley
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Claudio E. Tatsui
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA,Corresponding Author Claudio E. Tatsui Department of Neurosurgery, MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd, Houston, TX, USA
| |
Collapse
|
53
|
Hooiveld-Noeken JS, Eggen AC, Rácz E, de Vries EG, Reyners AK, Jalving M. Towards less mutilating treatments in patients with advanced non-melanoma skin cancers by earlier use of immune checkpoint inhibitors. Crit Rev Oncol Hematol 2022; 180:103855. [DOI: 10.1016/j.critrevonc.2022.103855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/24/2022] Open
|
54
|
Corrie P, Meyer N, Berardi R, Guidoboni M, Schlueter M, Kolovos S, Macabeo B, Trouiller JB, Laramée P. Comparative efficacy and safety of targeted therapies for BRAF-mutant unresectable or metastatic melanoma: Results from a systematic literature review and a network meta-analysis. Cancer Treat Rev 2022; 110:102463. [PMID: 36099854 DOI: 10.1016/j.ctrv.2022.102463] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The objective of this study was to estimate the relative efficacy and safety of targeted therapies for the treatment of metastatic melanoma using a network meta-analysis (NMA). METHODS A systematic literature review (SLR) identified studies in Medline, Embase and Cochrane published until November 2020. Screening used prespecified eligibility criteria. Following a transitivity assessment across included studies, Bayesian NMA was conducted. RESULTS A total of 43 publications reporting 15 targeted therapy trials and 42 reporting 18 immunotherapy trials were retained from the SLR and considered for the NMA. Due to substantial between-study heterogeneity with immunotherapy trials, the analysis considered a network restricted to targeted therapies. Among combination therapies, encorafenib + binimetinib was superior to dabrafenib + trametinib for overall response rate (OR = 1.86; 95 % credible interval [CrI] 1.10, 3.17), superior to vemurafenib + cobimetinib with fewer serious adverse events (SAEs) (OR = 0.51; 95 % CrI 0.29, 0.91) and fewer discontinuations due to AEs (OR = 0.45; 95 % CrI 0.21, 0.96), and superior to atezolizumab + vemurafenib + cobimetinib with fewer SAEs (OR = 0.41; 95 % CrI 0.21, 0.82). Atezolizumab + vemurafenib + cobimetinib and encorafenib + binimetinib were generally comparable for efficacy endpoints. Among double combination therapies, encorafenib + binimetinib showed high probabilities of being better for all efficacy and safety endpoints. CONCLUSIONS This NMA confirms that combination therapies are more efficacious than monotherapies. Encorafenib + binimetinib has a favourable efficacy profile compared to other double combination therapies and a favourable safety profile compared to both double and triple combination therapies.
Collapse
Affiliation(s)
- Pippa Corrie
- Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Nicolas Meyer
- Institut Universitaire du Cancer et CHU de Toulouse, Toulouse, France; Inserm UMR 1037 - CRCT, Toulouse, France
| | - Rossana Berardi
- Clinica Oncologica, AOU Ospedali Riuniti, Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Massimo Guidoboni
- Experimental and Clinical Oncology of Immunotherapy and Rare Tumors, IRCCS IRST "Dino Amadori", Meldola, FC, Italy
| | | | | | - Bérengère Macabeo
- Aix-Marseille Université, Marseille, France; Pierre Fabre Laboratories, Paris, France
| | | | - Philippe Laramée
- Aix-Marseille Université, Marseille, France; Pierre Fabre Laboratories, Paris, France.
| |
Collapse
|
55
|
Fontes SS, Nogueira ML, Dias RB, Rocha CAG, Soares MBP, Vannier-Santos MA, Bezerra DP. Combination Therapy of Curcumin and Disulfiram Synergistically Inhibits the Growth of B16-F10 Melanoma Cells by Inducing Oxidative Stress. Biomolecules 2022; 12:1600. [PMID: 36358950 PMCID: PMC9687191 DOI: 10.3390/biom12111600] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/11/2022] [Accepted: 10/16/2022] [Indexed: 10/05/2023] Open
Abstract
Oxidative stress plays a central role in the pathophysiology of melanoma. Curcumin (CUR) is a polyphenolic phytochemical that stimulates reactive oxygen species (ROS) production, while disulfiram (DSS) is a US FDA-approved drug for the treatment of alcoholism that can act by inhibiting the intracellular antioxidant system. Therefore, we hypothesized that they act synergistically against melanoma cells. Herein, we aimed to study the antitumor potential of the combination of CUR with DSS in B16-F10 melanoma cells using in vitro and in vivo models. The cytotoxic effects of different combination ratios of CUR and DSS were evaluated using the Alamar Blue method, allowing the production of isobolograms. Apoptosis detection, DNA fragmentation, cell cycle distribution, and mitochondrial superoxide levels were quantified by flow cytometry. Tumor development in vivo was evaluated using C57BL/6 mice bearing B16-F10 cells. The combinations ratios of 1:2, 1:3, and 2:3 showed synergic effects. B16-F10 cells treated with these combinations showed improved apoptotic cell death and DNA fragmentation. Enhanced mitochondrial superoxide levels were observed at combination ratios of 1:2 and 1:3, indicating increased oxidative stress. In vivo tumor growth inhibition for CUR (20 mg/kg), DSS (60 mg/kg), and their combination were 17.0%, 19.8%, and 28.8%, respectively. This study provided data on the potential cytotoxic activity of the combination of CUR with DSS and may provide a useful tool for the development of a therapeutic combination against melanoma.
Collapse
Affiliation(s)
- Sheila S. Fontes
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| | - Mateus L. Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| | - Rosane B. Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-909, BA, Brazil
| | - Clarissa A. Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-909, BA, Brazil
| | - Milena B. P. Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador 41650-010, BA, Brazil
| | | | - Daniel P. Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| |
Collapse
|
56
|
Signaling pathways and targeted therapies in lung squamous cell carcinoma: mechanisms and clinical trials. Signal Transduct Target Ther 2022; 7:353. [PMID: 36198685 PMCID: PMC9535022 DOI: 10.1038/s41392-022-01200-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/03/2022] [Accepted: 09/18/2022] [Indexed: 11/08/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death across the world. Unlike lung adenocarcinoma, patients with lung squamous cell carcinoma (LSCC) have not benefitted from targeted therapies. Although immunotherapy has significantly improved cancer patients' outcomes, the relatively low response rate and severe adverse events hinder the clinical application of this promising treatment in LSCC. Therefore, it is of vital importance to have a better understanding of the mechanisms underlying the pathogenesis of LSCC as well as the inner connection among different signaling pathways, which will surely provide opportunities for more effective therapeutic interventions for LSCC. In this review, new insights were given about classical signaling pathways which have been proved in other cancer types but not in LSCC, including PI3K signaling pathway, VEGF/VEGFR signaling, and CDK4/6 pathway. Other signaling pathways which may have therapeutic potentials in LSCC were also discussed, including the FGFR1 pathway, EGFR pathway, and KEAP1/NRF2 pathway. Next, chromosome 3q, which harbors two key squamous differentiation markers SOX2 and TP63 is discussed as well as its related potential therapeutic targets. We also provided some progress of LSCC in epigenetic therapies and immune checkpoints blockade (ICB) therapies. Subsequently, we outlined some combination strategies of ICB therapies and other targeted therapies. Finally, prospects and challenges were given related to the exploration and application of novel therapeutic strategies for LSCC.
Collapse
|
57
|
Bhattarai RS, Bariwal J, Kumar V, Hao C, Deng S, Li W, Mahato RI. pH-sensitive nanomedicine of novel tubulin polymerization inhibitor for lung metastatic melanoma. J Control Release 2022; 350:569-583. [PMID: 36037976 PMCID: PMC10322201 DOI: 10.1016/j.jconrel.2022.08.023] [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: 04/19/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 10/14/2022]
Abstract
Microtubule binding agents such as paclitaxel and vincristine have activity in metastatic melanoma. However, even responsive tumors develop resistance, highlighting the need to investigate new drug molecules. Here, we showed that a new compound, CH-2-102, developed by our group, has high anti-tumor efficacy in human and murine melanoma cells. We confirmed that CH-2-102 robustly suppresses the microtubule polymerization process by directly interacting with the colchicine binding site. Our results unveil that CH-2-102 suppresses microtubule polymerization and subsequently induces G2 phase cell arrest as one of the possible mechanisms. Notably, CH-2-102 maintains its efficacy even in the paclitaxel resistance melanoma cells due to different binding sites and a non-Pgp substrate. We developed a pH-responsive drug-polymer Schiff bases linker for high drug loading into nanoparticles (NPs). Our CH-2-102 conjugated NPs induced tumor regression more effectively than Abraxane® (Nab-paclitaxel, N-PTX), free drug, and non-sensitive NPs in B16-F10 cell-derived lung metastasis mouse model. Furthermore, our results suggest that the formulation has a high impact on the in vivo efficacy of the drug and warrants further investigation in other cancers, particularly taxane resistant. In conclusion, the microtubule polymerization inhibitor CH-2-102 conjugated pH-responsive NPs induce tumor regression in lung metastasis melanoma mice, suggesting it may be an effective strategy for treating metastatic melanoma.
Collapse
Affiliation(s)
- Rajan S Bhattarai
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jitender Bariwal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chen Hao
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
58
|
Rashid S, Shaughnessy M, Tsao H. Melanoma classification and management in the era of molecular medicine. Dermatol Clin 2022; 41:49-63. [DOI: 10.1016/j.det.2022.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
59
|
Lin EPY, Hsu CY, Berry L, Bunn P, Shyr Y. Analysis of Cancer Survival Associated With Immune Checkpoint Inhibitors After Statistical Adjustment: A Systematic Review and Meta-analyses. JAMA Netw Open 2022; 5:e2227211. [PMID: 35976648 PMCID: PMC9386543 DOI: 10.1001/jamanetworkopen.2022.27211] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
IMPORTANCE Appropriate clinical decision-making relies on accurate data interpretation, which in turn relies on the use of suitable statistical models. Long tails and early crossover-2 features commonly observed in immune checkpoint inhibitor (ICI) survival curves-raise questions as to the suitability of Cox proportional hazards regression for ICI survival analysis. Cox proportional hazards-Taylor expansion adjustment for long-term survival data (Cox-TEL) adjustment may provide possible solutions in this setting. OBJECTIVE To estimate overall survival and progression-free survival benefits of ICI therapy vs chemotherapy using Cox-TEL adjustment. DATA SOURCES A PubMed search was performed for all cataloged publications through May 22, 2022. STUDY SELECTION The search was restricted to randomized clinical trials with search terms for ICIs and lung cancer, melanoma, or urothelial carcinoma. The publications identified were further reviewed for inclusion. DATA EXTRACTION AND SYNTHESIS Cox proportional hazards ratios (HRs) were transformed to Cox-TEL HRs for patients with short-term treatment response (ie, short-term survivor) (ST-HR) and difference in proportions for patients with long-term survival (LT-DP) by Cox-TEL. Meta-analyses were performed using a frequentist random-effects model. MAIN OUTCOMES AND MEASURES Outcomes of interest were pooled overall survival (primary outcome) and progression-free survival (secondary outcome) HRs, ST-HRs, and LT-DPs. Subgroup analyses stratified by cancer type also were performed. RESULTS A total of 1036 publications was identified. After 3 levels of review against inclusion criteria, 13 clinical trials (7 in non-small cell lung cancer, 3 in melanoma, and 3 in urothelial carcinoma) were selected for the meta-analysis. In the primary analysis, pooled findings were 0.75 (95% CI, 0.70-0.81) for HR, 0.86 (95% CI, 0.81-0.92) for ST-HR, and 0.08 (95% CI, 0.06-0.10) for LT-DP. In the secondary analysis, the pooled values for progression-free survival were 0.77 (95% CI, 0.64-0.91) for HR, 1.02 (95% CI, 0.84-1.24) for ST-HR, and 0.10 (95% CI, 0.06-0.14) for LT-DP. CONCLUSIONS AND RELEVANCE This systematic review and meta-analysis of ICI clinical trial results noted consistently larger ST-HRs vs Cox HRs for ICI therapy, with an LT-DP of approximately 10%. These results suggest that Cox HRs may not provide a full picture of survival outcomes when the risk reduction from treatment is not constant, which may aid in the decision-making process of oncologists and patients.
Collapse
Affiliation(s)
- Emily Pei-Ying Lin
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Yuan Hsu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne Berry
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paul Bunn
- Department of Medicine, University of Colorado School of Medicine, Aurora
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
60
|
Pan W, Scherlinger M, Yoshida N, Tsokos MG, Tsokos GC. PPP2R2D Suppresses Effector T Cell Exhaustion and Regulatory T Cell Expansion and Inhibits Tumor Growth in Melanoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:621-628. [PMID: 35831019 PMCID: PMC9339485 DOI: 10.4049/jimmunol.2200041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/26/2022] [Indexed: 01/04/2023]
Abstract
We had shown previously that the protein phosphatase 2A regulatory subunit PPP2R2D suppresses IL-2 production, and PPP2R2D deficiency in T cells potentiates the suppressive function of regulatory T (Treg) cells and alleviates imiquimod-induced lupus-like pathology. In this study, in a melanoma xenograft model, we noted that the tumor grew in larger sizes in mice lacking PPP2R2D in T cells (LckCreR2Dfl/fl) compared with wild type (R2Dfl/fl) mice. The numbers of intratumoral T cells in LckCreR2Dfl/fl mice were reduced compared with R2Dfl/fl mice, and they expressed a PD-1+CD3+CD44+ exhaustion phenotype. In vitro experiments confirmed that the chromatin of exhaustion markers PD-1, LAG3, TIM3, and CTLA4 remained open in LckCreR2Dfl/fl CD4 T conventional compared with R2Dfl/fl T conventional cells. Moreover, the percentage of Treg cells (CD3+CD4+Foxp3+CD25hi) was significantly increased in the xenografted tumor of LckCreR2Dfl/fl mice compared with R2Dfl/fl mice probably because of the increase in the percentage of IL-2-producing LckCreR2Dfl/fl T cells. Moreover, using adoptive T cell transfer in mice xenografted with melanoma, we demonstrated that PPP2R2D deficiency in T cells enhanced the inhibitory effect of Treg cells in antitumor immunity. At the translational level, analysis of publicly available data from 418 patients with melanoma revealed that PPP2R2D expression levels correlated positively with tumor-infiltration level of CD4 and CD8 T cells. The data demonstrate that PPP2R2D is a negative regulator of immune checkpoint receptors, and its absence exacerbates effector T cell exhaustion and promotes Treg cell expansion. We conclude that PPP2R2D protects against melanoma growth, and PPP2R2D-promoting regimens can have therapeutic value in patients with melanoma.
Collapse
Affiliation(s)
- Wenliang Pan
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Marc Scherlinger
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
61
|
Chokeberry (Aronia melanocarpa) fruit extract abrogates melanoma progression through boosting up IFN-γ-producing cells. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
62
|
Song Y, Huang J, Wang K, Li Y. To Identify Adenomatous Polyposis Coli Gene Mutation as a Predictive Marker of Endometrial Cancer Immunotherapy. Front Cell Dev Biol 2022; 10:935650. [PMID: 35938175 PMCID: PMC9354690 DOI: 10.3389/fcell.2022.935650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
The adenomatous polyposis coli (APC) gene is the chromatin-remodeling-related gene and a typical tumor suppressor. Patients with a high expression of programmed death-ligand 1 (PD-L1) or a high level of tumor mutational burden (TMB) may benefit from immunotherapy in endometrial cancer (EC). This study aimed to demonstrate the role of APC in the diagnosis and immunotherapy treatment of EC. We performed an integrative analysis of a commercial panel including 520 cancer-related genes on 99 tumors from an endometrial cancer cohort in China and DNA-seq data from The Cancer Genome Atlas (TCGA) to identify new gene mutations as endometrial cancer immunotherapy markers. We found that the significant mutant genes that correlated with the PD-L1 expression and TMB were related to the chromatin state and generated a discovery set having 12 mutated genes, including the APC gene, which was identified as a new marker for immunotherapy. Further analysis revealed that tumors with the APC mutation had high TMB, increased expression of PD-L1, and increased lymphocytic infiltration. Next, we verified that APC has an inactive mutation in EC, which may affect the immune response, including PD-L1 expression, microsatellite instability, and lymphocytic infiltrate. Furthermore, patients with the APC mutation had longer overall survival. Our study demonstrates that APC could play an important role in enhancing the response to endometrial cancer treatment, particularly immunotherapy.
Collapse
Affiliation(s)
- Yunfeng Song
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Huang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Kai Wang, ; Yiran Li,
| | - Yiran Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
- Harvard Medical School, Boston, MA, United States
- *Correspondence: Kai Wang, ; Yiran Li,
| |
Collapse
|
63
|
Proliferation and Immune Response Gene Signatures Associated with Clinical Outcome to Immunotherapy and Targeted Therapy in Metastatic Cutaneous Malignant Melanoma. Cancers (Basel) 2022; 14:cancers14153587. [PMID: 35892846 PMCID: PMC9331037 DOI: 10.3390/cancers14153587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/07/2022] [Accepted: 07/13/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary The introduction of treatment with targeted therapies and immunotherapies has dramatically changed the outcome for patients with advanced cutaneous melanoma. However, only a subset of the patients has durable benefits from the treatment. This exploratory study aims to identify genes/gene signatures as predictive biomarkers for treatment outcomes in melanoma. Targeted transcriptomics and gene set enrichment analysis (GSEA) were applied in 28 melanoma samples collected before receiving treatment. Thirteen patients were treated with targeted therapy (TT) and 15 patients were treated with immune checkpoint inhibitors (ICI). Up-regulation of genes involved in immune processes was associated with a better outcome of TT. Down-regulation of proliferation and up-regulation of allograft rejection gene sets favored ICI patients. Further follow-up of the inverse relation between proliferation and allograft rejection gene signatures and relation to outcome is warranted. Abstract Targeted therapy (TT), together with immune checkpoint inhibitors (ICI), has significantly improved clinical outcomes for patients with advanced cutaneous malignant melanoma (CMM) during the last decade. However, the magnitude and the duration of response vary considerably. There is still a paucity of predictive biomarkers to identify patients who benefit most from treatment. To address this, we performed targeted transcriptomics of CMM tumors to identify biomarkers associated with clinical outcomes. Pre-treatment tumor samples from 28 patients with advanced CMM receiving TT (n = 13) or ICI (n = 15) were included in the study. Targeted RNA sequencing was performed using Ion AmpliSeq ™, followed by gene set enrichment analysis (GSEA) using MSigDB’s Hallmark Gene Set Collection to identify gene expression signatures correlating with treatment outcome. The GSEA demonstrated that up-regulation of allograft rejection genes, together with down-regulation of E2F and MYC targets as well as G2M checkpoint genes, significantly correlated with longer progression-free survival on ICI while IFNγ and inflammatory response genes were associated with a better clinical outcome on TT. In conclusion, we identify novel genes and their expression signatures as potential predictive biomarkers for TT and ICI in patients with metastatic CMM, paving the way for clinical use following larger validation studies.
Collapse
|
64
|
Jing M, Cai Y, Shi J, Zhang X, Zhu B, Yuan F, Zhang J, Xiao M, Chen M. Adjuvant Treatments of Adult Melanoma: A Systematic Review and Network Meta-Analysis. Front Oncol 2022; 12:926242. [PMID: 35785213 PMCID: PMC9247312 DOI: 10.3389/fonc.2022.926242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
Multiple treatments of unresectable advanced or metastatic melanoma have been licensed in the adjuvant setting, causing tremendous interest in developing neoadjuvant strategies for melanoma. Eligible studies included those that compared overall survival/progression-free survival/grade 3 or 4 adverse events in patients with unresectable advanced or metastatic melanoma. Seven eligible randomized trials with nine publications were included in this study. Direct and network meta-analysis consistently indicated that nivolumab+ipilimumab, nivolumab, and trametinib could significantly improve overall survival and progression-free survival compared to ipilimumab in advanced melanoma patients. Compared to ipilimumab, nivolumab, dacarbazine, and ipilimumab+gp100 had a reduced risk of grade 3/4 adverse reactions. The nivolumab+ipilimumab combination had the highest risk of adverse events, followed by ipilimumab+dacarbazine and trametinib. Combination therapy was more beneficial to improve overall survival and progression-free survival than monotherapy in advanced melanoma treatment, albeit at the cost of increased toxicity. Regarding the overall survival/progression-free survival, ipilimumab+gp100 ranked below ipilimumab+dacarbazine and nivolumab+ipilimumab, although it had a smaller rate of grade 3 or 4 AEs than other treatments (except nivolumab). Nivolumab is the optimum adjuvant treatment for unresectable advanced or metastatic melanoma with a good risk-benefit profile. In order to choose the best therapy, clinicians must consider the efficacy, adverse events, and physical status.
Collapse
Affiliation(s)
- Mingyi Jing
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Cai
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Shi
- Department of Internal Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xufan Zhang
- Department of Nuclear Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Baohua Zhu
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fan Yuan
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Urology & Andrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhang
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Xiao
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Min Xiao, ; Mingling Chen,
| | - Mingling Chen
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Min Xiao, ; Mingling Chen,
| |
Collapse
|
65
|
Gorris MAJ, van der Woude LL, Kroeze LI, Bol K, Verrijp K, Amir AL, Meek J, Textor J, Figdor CG, de Vries IJM. Paired primary and metastatic lesions of patients with ipilimumab-treated melanoma: high variation in lymphocyte infiltration and HLA-ABC expression whereas tumor mutational load is similar and correlates with clinical outcome. J Immunother Cancer 2022; 10:e004329. [PMID: 35550553 PMCID: PMC9109111 DOI: 10.1136/jitc-2021-004329] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) can lead to long-term responses in patients with metastatic melanoma. Still many patients with melanoma are intrinsically resistant or acquire secondary resistance. Previous studies have used primary or metastatic tumor tissue for biomarker assessment. Especially in melanoma, metastatic lesions are often present at different anatomical sites such as skin, lymph nodes, and visceral organs. The anatomical site may directly affect the tumor microenvironment (TME). To evaluate the impact of tumor evolution on the TME and on ICI treatment outcome, we directly compared paired primary and metastatic melanoma lesions for tumor mutational burden (TMB), HLA-ABC status, and tumor infiltrating lymphocytes (TILs) of patients that received ipilimumab. METHODS TMB was analyzed by sequencing primary and metastatic melanoma lesions using the TruSight Oncology 500 assay. Tumor tissues were subjected to multiplex immunohistochemistry to assess HLA-ABC status and for the detection of TIL subsets (B cells, cytotoxic T cells, helper T cells, and regulatory T cells), by using a machine-learning algorithm. RESULTS While we observed a very good agreement between TMB of matched primary and metastatic melanoma lesions (intraclass coefficient=0.921), such association was absent for HLA-ABC status, TIL density, and subsets thereof. Interestingly, analyses of different metastatic melanoma lesions within a single patient revealed that TIL density and composition agreed remarkably well, rejecting the hypothesis that the TME of different anatomical sites affects TIL infiltration. Similarly, the HLA-ABC status between different metastatic lesions within patients was also comparable. Furthermore, high TMB, of either primary or metastatic melanoma tissue, directly correlated with response to ipilimumab, whereas lymphocyte density or composition did not. Loss of HLA-ABC in the metastatic lesion correlated to a shorter progression-free survival on ipilimumab. CONCLUSIONS We confirm the link between TMB and HLA-ABC status and the response to ipilimumab-based immunotherapy in melanoma, but no correlation was found for TIL density, neither in primary nor metastatic lesions. Our finding that TMB between paired primary and metastatic melanoma lesions is highly stable, demonstrates its independency of the time point and location of acquisition. TIL and HLA-ABC status in metastatic lesions of different anatomical sites are highly similar within an individual patient.
Collapse
Affiliation(s)
- Mark A J Gorris
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
| | - Lieke L van der Woude
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
- Pathology, Radboudumc, Nijmegen, The Netherlands
| | | | - Kalijn Bol
- Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Oncode Institute, Nijmegen, The Netherlands
- Pathology, Radboudumc, Nijmegen, The Netherlands
| | | | - Jelena Meek
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Data Science Group, Institute for Computing and Information Sciences, Radboud Universiteit, Nijmegen, The Netherlands
| | - Carl G Figdor
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
| | | |
Collapse
|
66
|
van Luijk IF, Smith SM, Marte Ojeda MC, Oei AL, Kenter GG, Jordanova ES. A Review of the Effects of Cervical Cancer Standard Treatment on Immune Parameters in Peripheral Blood, Tumor Draining Lymph Nodes, and Local Tumor Microenvironment. J Clin Med 2022; 11:2277. [PMID: 35566403 PMCID: PMC9102821 DOI: 10.3390/jcm11092277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Cervical cancer remains a public health concern despite all the efforts to implement vaccination and screening programs. Conventional treatment for locally advanced cervical cancer consists of surgery, radiotherapy (with concurrent brachytherapy), combined with chemotherapy, or hyperthermia. The response rate to combination approaches involving immunomodulatory agents and conventional treatment modalities have been explored but remain dismal in patients with locally advanced disease. Studies exploring the immunological effects exerted by combination treatment modalities at the different levels of the immune system (peripheral blood (PB), tumor-draining lymph nodes (TDLN), and the local tumor microenvironment (TME)) are scarce. In this systemic review, we aim to define immunomodulatory and immunosuppressive effects induced by conventional treatment in cervical cancer patients to identify the optimal time point for immunotherapy administration. Radiotherapy (RT) and chemoradiation (CRT) induce an immunosuppressive state characterized by a long-lasting reduction in peripheral CD3, CD4, CD8 T cells and NK cells. At the TDLN level, CRT induced a reduction in Nrp1+Treg stability and number, naïve CD4 and CD8 T cell numbers, and an accompanying increase in IFNγ-producing CD4 helper T cells, CD8 T cells, and NK cells. Potentiation of the T-cell anti-tumor response was particularly observed in patients receiving low irradiation dosage. At the level of the TME, CRT induced a rebound effect characterized by a reduction of the T-cell anti-tumor response followed by stable radioresistant OX40 and FoxP3 Treg cell numbers. However, the effects induced by CRT were very heterogeneous across studies. Neoadjuvant chemotherapy (NACT) containing both paclitaxel and cisplatin induced a reduction in stromal FoxP3 Treg numbers and an increase in stromal and intratumoral CD8 T cells. Both CRT and NACT induced an increase in PD-L1 expression. Although there was no association between pre-treatment PD-L1 expression and treatment outcome, the data hint at an association with pro-inflammatory immune signatures, overall and disease-specific survival (OS, DSS). When considering NACT, we propose that posterior immunotherapy might further reduce immunosuppression and chemoresistance. This review points at differential effects induced by conventional treatment modalities at different immune compartments, thus, the compartmentalization of the immune responses as well as individual patient's treatment plans should be carefully considered when designing immunotherapy treatment regimens.
Collapse
Affiliation(s)
- Iske F. van Luijk
- Haaglanden Medical Center, Lijnbaan 32, 2512 VA The Hague, The Netherlands
- Center for Gynecologic Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.C.M.O.); (G.G.K.); (E.S.J.)
| | - Sharissa M. Smith
- Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Maria C. Marte Ojeda
- Center for Gynecologic Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.C.M.O.); (G.G.K.); (E.S.J.)
| | - Arlene L. Oei
- Laboratory for Experimental Oncology and Radiobiology, Department of Radiation Oncology, Amsterdam UMC, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Gemma G. Kenter
- Center for Gynecologic Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.C.M.O.); (G.G.K.); (E.S.J.)
| | - Ekaterina S. Jordanova
- Center for Gynecologic Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.C.M.O.); (G.G.K.); (E.S.J.)
- Department of Urology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
67
|
Mercier R, LaPointe P. The role of cellular proteostasis in anti-tumor immunity. J Biol Chem 2022; 298:101930. [PMID: 35421375 PMCID: PMC9108985 DOI: 10.1016/j.jbc.2022.101930] [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: 02/02/2022] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/25/2022] Open
Abstract
Immune checkpoint blockade therapy is perhaps the most important development in cancer treatment in recent memory. It is based on decades of investigation into the biology of immune cells and the role of the immune system in controlling cancer growth. While the molecular circuitry that governs the immune system in general - and anti-tumor immunity in particular - is intensely studied, far less attention has been paid to the role of cellular stress in this process. Proteostasis, intimately linked to cell stress responses, refers to the dynamic regulation of the cellular proteome and is maintained through a complex network of systems that govern the synthesis, folding, and degradation of proteins in the cell. Disruption of these systems can result in the loss of protein function, altered protein function, the formation of toxic aggregates, or pathologies associated with cell stress. However, the importance of proteostasis extends beyond its role in maintaining proper protein function; proteostasis governs how tolerant cells may be to mutations in protein coding genes and the overall half-life of proteins. Such gene expression changes may be associated with human diseases including neurodegenerative diseases, metabolic disease, and cancer and manifest at the protein level against the backdrop of the proteostasis network in any given cellular environment. In this review, we focus on the role of proteostasis in regulating immune responses against cancer as well the role of proteostasis in determining immunogenicity of cancer cells.
Collapse
Affiliation(s)
- Rebecca Mercier
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Paul LaPointe
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
68
|
Sabbatino F, Liguori L, Pepe S, Ferrone S. Immune checkpoint inhibitors for the treatment of melanoma. Expert Opin Biol Ther 2022; 22:563-576. [PMID: 35130816 PMCID: PMC9038682 DOI: 10.1080/14712598.2022.2038132] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Immune checkpoint inhibitor (ICI) based immunotherapy is dramatically changing the management of many types of cancers including melanoma. In this malignancy, ICIs have been shown to prolong disease and progression free survival as well as overall survival of a percentage of treated patients, becoming the cornerstone of melanoma treatment. AREAS COVERED : In this review, first, we will describe the mechanisms of immune checkpoint activation and inhibition, second, we will summarize the results obtained with ICIs in melanoma treatment in terms of efficacy as well as toxicity, third, we will discuss the potential mechanisms of immune escape from ICI, and lastly, we will review the potential predictive biomarkers of clinical efficacy of ICI-based immunotherapy in melanoma. EXPERT OPINION : ICIs represent one of the pillars of melanoma treatment. The success of ICI-based therapy is limited by the development of escape mechanisms which allow melanoma cells to avoid recognition and destruction by immune cells. These results emphasize the need of additional studies to confirm the efficacy of therapies which combine different classes of ICIs as well as ICIs with other types of therapies. Furthermore, novel and more effective predictive biomarkers are needed to better stratify melanoma patients in order to define more precisely the therapeutic algorithms.
Collapse
Affiliation(s)
- Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy 84131
| | - Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy 80131
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy 84131
| | - Soldano Ferrone
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
69
|
Tong S, Cinelli MA, El-Sayed NS, Huang H, Patel A, Silverman RB, Yang S. Inhibition of interferon-gamma-stimulated melanoma progression by targeting neuronal nitric oxide synthase (nNOS). Sci Rep 2022; 12:1701. [PMID: 35105915 PMCID: PMC8807785 DOI: 10.1038/s41598-022-05394-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023] Open
Abstract
Interferon-gamma (IFN-γ) is shown to stimulate melanoma development and progression. However, the underlying mechanism has not been completely defined. Our study aimed to determine the role of neuronal nitric oxide synthase (nNOS)-mediated signaling in IFN-γ-stimulated melanoma progression and the anti-melanoma effects of novel nNOS inhibitors. Our study shows that IFN-γ markedly induced the expression levels of nNOS in melanoma cells associated with increased intracellular nitric oxide (NO) levels. Co-treatment with novel nNOS inhibitors effectively alleviated IFN-γ-activated STAT1/3. Further, reverse phase protein array (RPPA) analysis demonstrated that IFN-γ induced the expression of HIF1α, c-Myc, and programmed death-ligand 1 (PD-L1), in contrast to IFN-α. Blocking the nNOS-mediated signaling pathway using nNOS-selective inhibitors was shown to effectively diminish IFN-γ-induced PD-L1 expression in melanoma cells. Using a human melanoma xenograft mouse model, the in vivo studies revealed that IFN-γ increased tumor growth compared to control, which was inhibited by the co-administration of nNOS inhibitor MAC-3-190. Another nNOS inhibitor, HH044, was shown to effectively inhibit in vivo tumor growth and was associated with reduced PD-L1 expression levels in melanoma xenografts. Our study demonstrates the important role of nNOS-mediated NO signaling in IFN-γ-stimulated melanoma progression. Targeting nNOS using highly selective small molecular inhibitors is a unique and effective strategy to improve melanoma treatment.
Collapse
Affiliation(s)
- Shirley Tong
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, #297-Y, 9401 Jeronimo Road, Irvine, CA, 92618, USA
| | - Maris A Cinelli
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL, 60208, USA
| | - Naglaa Salem El-Sayed
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, #297-Y, 9401 Jeronimo Road, Irvine, CA, 92618, USA
| | - He Huang
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL, 60208, USA
| | - Anika Patel
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, #297-Y, 9401 Jeronimo Road, Irvine, CA, 92618, USA
| | - Richard B Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL, 60208, USA.,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sun Yang
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, #297-Y, 9401 Jeronimo Road, Irvine, CA, 92618, USA.
| |
Collapse
|
70
|
Graziani G, Lisi L, Tentori L, Navarra P. Monoclonal Antibodies to CTLA-4 with Focus on Ipilimumab. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:295-350. [PMID: 35165868 DOI: 10.1007/978-3-030-91311-3_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The immune checkpoint cytotoxic T lymphocyte-associated antigen 4 (CTLA-4 or CD152) is a negative regulator of T-cell-mediated immune responses which plays a critical role in suppressing autoimmunity and maintaining immune homeostasis. Because of its inhibitory activity on T cells, CTLA-4 has been investigated as a drug target to induce immunostimulation, blocking the interaction with its ligands. The antitumor effects mediated by CTLA-4 blockade have been attributed to a sustained active immune response against cancer cells, due to the release of a brake on T cell activation. Ipilimumab (Yervoy, Bristol-Myers Squibb) is a fully human anti-CTLA-4 IgG1κ monoclonal antibody (mAb) that represents the first immune checkpoint inhibitor approved as monotherapy by FDA and EMA in 2011 for the treatment of unresectable/metastatic melanoma. In 2015, FDA also granted approval to ipilimumab monotherapy as adjuvant treatment of stage III melanoma to reduce the risk of tumour recurrence. The subsequent approved indications of ipilimumab for metastatic melanoma, regardless of BRAF mutational status, and other advanced/metastatic solid tumours always involve its use in association with the anti-programmed cell death protein 1 (PD-1) mAb nivolumab. Currently, ipilimumab is evaluated in ongoing clinical trials for refractory/advanced solid tumours mainly in combination with additional immunostimulating agents.
Collapse
Affiliation(s)
- Grazia Graziani
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Lucio Tentori
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
71
|
Schweighofer-Zwink G, Manafi-Farid R, Kölblinger P, Hehenwarter L, Harsini S, Pirich C, Beheshti M. Prognostic value of 2-[ 18F]FDG PET-CT in metastatic melanoma patients receiving immunotherapy. Eur J Radiol 2021; 146:110107. [PMID: 34922117 DOI: 10.1016/j.ejrad.2021.110107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/10/2021] [Accepted: 12/08/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE The 2-fluorodeoxyglucose positron emission tomography/computed tomography (2-[18F]FDG PET/CT) is used for the evaluation of response to immunotherapy in malignant melanoma. Here, we evaluated the prognostic value of various metabolic parameters in baseline and different time points after therapy. METHODS In this retrospective study, 51 metastatic melanoma patients, who had received immunotherapy, were included. Patients with baseline and two follow-up 2-[18F]FDG PET/CT studies (3 and 6 months after therapy) were selected. Multiple metabolic parameters and tumor-to-background ratios (TBRs) were extracted and correlated with OS. RESULTS The 3- and 5-year OS rates were 49% and 43.1%, respectively. On baseline 2-[18F]FDG PET/CT, only standardized uptake value corrected for lean body mass (SULmax and SULpeak), as well as most of the TBRs were predictive for 3- and 5-year OS rates. Metabolic tumor volume (MTV), total lesion glycolysis (TLG), and most of the TBRs were predictive on both follow-up studies. Also, the changes in values of MTV, TLG and most of the TBRs from the baseline to the 3-month and 6- month follow-up studies were prognostic. On multivariate analysis, all of the most predictive parameters for OS were derived from the 3-month follow-up study. The ratio of TBRmean to the mediastinum was the best factor (cutoff value of 2.15, sensitivity of 88.5% and specificity of 68.0% for 3-year survival). CONCLUSION Metabolic parameters derived from 2-[18F]FDG PET/CT are valuable tools for the prediction of 3- and 5-year OS rates in metastatic melanoma patients undergoing immunotherapy. The 3-month follow-up 2-[18F]FDG PET/CT is of particular importance in this regard.
Collapse
Affiliation(s)
- Gregor Schweighofer-Zwink
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical sciences, 1411713135 Tehran, Iran
| | - Peter Kölblinger
- Department of Dermatology, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Lukas Hehenwarter
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Sara Harsini
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical sciences, 1411713135 Tehran, Iran; Association of Nuclear Medicine and Molecular Imaging (ANMMI), Universal Scientific Education and Research Network (USERN), 1419733151 Tehran, Iran
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria.
| |
Collapse
|
72
|
Hughes T, Klairmont M, Sharfman WH, Kaufman HL. Interleukin-2, Ipilimumab, and Anti-PD-1: clinical management and the evolving role of immunotherapy for the treatment of patients with metastatic melanoma. Cancer Biol Ther 2021; 22:513-526. [PMID: 26418961 PMCID: PMC8726727 DOI: 10.1080/15384047.2015.1095401] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/10/2015] [Accepted: 09/12/2015] [Indexed: 12/20/2022] Open
Abstract
Treatment of metastatic melanoma has changed dramatically in the past 5 years with the approval of six new agents (vemurafenib, dabrafenib, trametinib, ipilimumab, pembrolizumab, and nivolumab) by the US Food and Drug Administration (FDA). This review will compare the immunotherapies recently approved by the FDA (ipilimumab, nivolumab and pembrolizumab) with the long-approved immunotherapy, interleukin-2. Additional consideration will be given to the evolving landscape, including the opportunities for combination regimens. Immunotherapies have distinct mechanisms of action and unique response kinetics that differ from conventional cytotoxic and targeted therapies, and have a range of adverse events that can be safely managed by experienced health-care providers. Data suggest immunotherapies can result in long-term survival in a proportion of patients. This dynamic and evolving field of immunotherapy for melanoma will continue to offer challenges in terms of optimal patient management for the foreseeable future.
Collapse
|
73
|
Chen X, Zhou M, Wang Z, Lu S, Chang S, Zhou Z. Immunotherapy treatment outcome prediction in metastatic melanoma through an automated multi-objective delta-radiomics model. Comput Biol Med 2021; 138:104916. [PMID: 34656867 DOI: 10.1016/j.compbiomed.2021.104916] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 01/18/2023]
Abstract
Based on recent studies, immunotherapy led by immune checkpoint inhibitors has significantly improved the patient survival rate and effectively reduced the recurrence risk. However, immunotherapy has different therapeutic effects for different patients, leading to difficulties in predicting the treatment response. Conversely, delta-radiomic features, which measure the difference between pre- and post-treatment through quantitative image features, have proven to be promising descriptors for treatment outcome prediction. Consequently, we developed an effective model termed as the automated multi-objective delta-radiomics (Auto-MODR) model for the prediction of immunotherapy response in metastatic melanoma. In Auto-MODR, delta-radiomic features and traditional radiomic features were used as inputs. Furthermore, a novel automated multi-objective model was developed to obtain more reliable and balanced results between sensitivity and specificity. We conducted extensive comparisons with existing studies on treatment outcome prediction. Our method achieved an area under the curve (AUC) of 0.86 in a cross-validation study and an AUC of 0.73 in an independent study. Compared with the model using conventional radiomic features (pre- and post-treatment) only, better performance can be obtained when conventional radiomic and delta-radiomic features are combined. Furthermore, Auto-MODR outperformed the currently available radiomic strategies.
Collapse
Affiliation(s)
- Xi Chen
- School of Information and Communication Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Meijuan Zhou
- School of Information and Communication Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Zhilong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Si Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shaojie Chang
- School of Information and Communication Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Zhiguo Zhou
- School of Computer Science and Mathematics, University of Central Missouri, Warrensburg, MO, USA.
| |
Collapse
|
74
|
Larroquette M, Domblides C, Lefort F, Lasserre M, Quivy A, Sionneau B, Bertolaso P, Gross-Goupil M, Ravaud A, Daste A. Combining immune checkpoint inhibitors with chemotherapy in advanced solid tumours: A review. Eur J Cancer 2021; 158:47-62. [PMID: 34655837 DOI: 10.1016/j.ejca.2021.09.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/11/2021] [Indexed: 12/30/2022]
Abstract
The use of immune checkpoint inhibitors (ICIs), especially anti-programmed cell death 1 (PD1) and anti-programmed cell death ligand 1 (PD-L1), has changed practices in oncology, becoming a new standard of care in first or subsequent lines for several cancer subtypes. Recent data have highlighted the ability of standard chemotherapy to enhance immunogenicity and/or to break immunoresistance of the tumour and its microenvironment, leading to a rationale for the use of ICIs in combination with the standard chemotherapy regimen to improve efficacy of cancer treatment. Here, we propose to review randomised clinical trials evaluating concomitant administration of ICIs and chemotherapy, to assess clinical efficacy and safety profiles in advanced solid tumours. Association of these two modes of action on treatments has shown improved overall survival and better objective response rates than standard chemotherapy, especially in first-line treatment of non-small cell lung cancer (NSCLC) and for PD1/PD-L1 enriched tumours, highlighting a potential synergistic effect of this treatment combination in certain tumour types. However, improved survival results with the use of anti-PD-L1 avelumab as a maintenance schedule for bladder cancer raises the question of the most appropriate approach between sequential and concomitant administration of chemoimmunotherapy. To date, no trials have compared in a head-to-head protocol the administration of concomitant chemoimmunotherapy with chemotherapy, used for tumour debulking, followed by administration of ICIs. Regarding the tolerance profile, no new safety signals were found with the combination tested to date. Interestingly, recent results have shown an improved Progression Free survival 2 (PFS2) (defined as the progression after the next line of therapy) in head-and-neck cancers or NSCLC after a first-line pembrolizumab-chemotherapy combination, suggesting a potential long-lasting effect of ICIs when used in combination in the first-line setting.
Collapse
Affiliation(s)
- Mathieu Larroquette
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France; Bordeaux University, Bordeaux, France
| | - Charlotte Domblides
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France; Bordeaux University, Bordeaux, France
| | - Félix Lefort
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France
| | - Matthieu Lasserre
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France; Bordeaux University, Bordeaux, France
| | - Amandine Quivy
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France
| | - Baptiste Sionneau
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France
| | - Pauline Bertolaso
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France
| | - Alain Ravaud
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France
| | - Amaury Daste
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, Bordeaux, France.
| |
Collapse
|
75
|
Shigdar S, Schrand B, Giangrande PH, de Franciscis V. Aptamers: Cutting edge of cancer therapies. Mol Ther 2021; 29:2396-2411. [PMID: 34146729 PMCID: PMC8353241 DOI: 10.1016/j.ymthe.2021.06.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
The development of an aptamer-based therapeutic has rapidly progressed following the first two reports in the 1990s, underscoring the advantages of aptamer drugs associated with their unique binding properties. In 2004, the US Food and Drug Administration (FDA) approved the first therapeutic aptamer for the treatment of neovascular age-related macular degeneration, Macugen developed by NeXstar. Since then, eleven aptamers have successfully entered clinical trials for various therapeutic indications. Despite some of the pre-clinical and clinical successes of aptamers as therapeutics, no aptamer has been approved by the FDA for the treatment of cancer. This review highlights the most recent and cutting-edge approaches in the development of aptamers for the treatment of cancer types most refractory to conventional therapies. Herein, we will review (1) the development of aptamers to enhance anti-cancer immunity and as delivery tools for inducing the expression of immunogenic neoantigens; (2) the development of the most promising therapeutic aptamers designed to target the hard-to-treat cancers such as brain tumors; and (3) the development of "carrier" aptamers able to target and penetrate tumors and metastasis, delivering RNA therapeutics to the cytosol and nucleus.
Collapse
Affiliation(s)
- Sarah Shigdar
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, 75 Pigdons Road, Waurn Ponds, VIC 3216, Australia
| | - Brett Schrand
- TCR(2) Therapeutics, Inc., 100 Binney Street, Cambridge, MA 02142, USA
| | - Paloma H Giangrande
- Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; VP Platform Discovery Sciences, Biology, Wave Life Sciences, Cambridge, MA 02138, USA
| | - Vittorio de Franciscis
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan, Italy; Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
76
|
Tully KH, Cone EB, Cole AP, Sun M, Chen X, Marchese M, Roghmann F, Kilbridge KL, Trinh QD. Risk of Immune-related Adverse Events in Melanoma Patients With Preexisting Autoimmune Disease Treated With Immune Checkpoint Inhibitors: A Population-based Study Using SEER-Medicare Data. Am J Clin Oncol 2021; 44:413-418. [PMID: 34081033 DOI: 10.1097/coc.0000000000000840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The objective of this study was to examine the risk of immune-related adverse events (irAEs) in patients with a preexisting autoimmune disease (pAID) presenting with a cutaneous melanoma receiving an immune checkpoint inhibitor (ICI) therapy. METHODS Data from the Surveillance, Epidemiology, and End Results cancer registries and linked Medicare claims between January 2010 and December 2015 was used to identify patients diagnosed with cutaneous melanoma who had pAID or received ICI or both. Patients were then stratified into 3 groups: ICI+pAID, non-ICI+pAID, and ICI+non-pAID. Inverse probability of treatment weighted Cox proportional hazards regression models were fitted to assess the risk of cardiac, pulmonary, endocrine, and neurological irAE. RESULTS In total, 3704 individuals were included in the analysis. The majority of patients consisted of non-ICI+pAID patients (N=2706/73.1%), while 106 (2.9%) patients and 892 (24.1%) were classified as ICI+pAID and ICI+non-pAID, respectively. The risk of irAE was higher in the ICI+pAID group compared with the non-ICI+pAID and ICI+non-pAID, respectively (non-ICI: cardiac: hazard ratio [HR]=3.59, 95% confidence interval [CI]: 2.83-4.55; pulmonary: HR=3.94, 95% CI: 3.23-4.81; endocrine: HR=1.72, 95% CI: 1.53-1.93; neurological: HR=3.88, 95% CI: 2.30-6.57/non-pAID: cardiac: HR=3.83, 95% CI: 3.39-4.32; pulmonary: HR=2.08, 95% CI: 1.87-2.32; endocrine: HR=1.23, 95% CI: 1.14-1.32; neurological: HR=3.77, 95% CI: 2.75-5.18). CONCLUSIONS Patients with a pAID face a significantly higher risk of irAEs. Further research examining the clinical impact of these events on the patients' oncological outcome and quality of life is urgently needed given our findings of significantly worse rates of adverse events.
Collapse
Affiliation(s)
- Karl H Tully
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School
- Department of Urology and Neurourology, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Eugene B Cone
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School
| | - Alexander P Cole
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School
| | - Maxine Sun
- Lank Center for Genitourinary Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA
| | - Xi Chen
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School
| | - Maya Marchese
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School
| | - Florian Roghmann
- Department of Urology and Neurourology, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Kerry L Kilbridge
- Department of Urology and Neurourology, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Quoc-Dien Trinh
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School
| |
Collapse
|
77
|
Bouchereau S, Chaplain L, Fort M, Beauchet A, Sidibé T, Chapalain M, Gonzalez-Lara L, Longvert C, Blom A, Saiag P, Funck-Brentano E. Impact of prior treatment with immune checkpoint inhibitors on dacarbazine efficacy in metastatic melanoma. Br J Cancer 2021; 125:948-954. [PMID: 34262147 DOI: 10.1038/s41416-021-01486-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/07/2021] [Accepted: 07/01/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite its low efficacy, chemotherapy with dacarbazine remains an option in metastatic melanoma patients after failure of immune checkpoint inhibitors (ICI) ± targeted therapy. Some observations suggested an increased efficacy of chemotherapy in melanoma or lung cancer patients previously treated with ICI; we aimed to evaluate the efficacy of dacarbazine in a controlled-group study of patients pre-treated or not with ICI. METHODS We retrospectively collected data from all consecutive patients treated with dacarbazine for advanced cutaneous melanoma without brain metastasis, in our skin cancer centre between June 2006 and September 2019. The primary endpoint was progression-free survival (PFS); secondary endpoints were overall response rates (ORR), overall survival (OS) and safety of dacarbazine. RESULTS Among 72 patients, 17 (23.6%) received dacarbazine after ICI and 55 (76.3%) without prior ICI. Despite less favourable prognostic factors in patients ICI-pre-treated, median PFS was 4.27 months (range 0.89-43.69) in this group versus 2.04 months (range 1.25-39.25) P = 0.03 in non-ICI-pre-treated patients; ORR were 35.3% and 12.7%, respectively. The median OS and the occurrence of adverse events were similar in both groups. CONCLUSION Dacarbazine seems to offer a short-lived benefit in patients with progressive advanced disease despite ICI (±targeted therapy), and could be an alternative before considering best supportive care.
Collapse
Affiliation(s)
- Sarah Bouchereau
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Louise Chaplain
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France.,Research unit EA4340 'Biomarkers and clinical trials in cancerology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, Boulogne-Billancourt, France
| | - Magali Fort
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Alain Beauchet
- Department of Public Health, GHU Paris-Saclay, AP-HP, Boulogne-Billancourt, France
| | - Thomas Sidibé
- Department of Pharmacy, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Marie Chapalain
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Leire Gonzalez-Lara
- Research unit EA4340 'Biomarkers and clinical trials in cancerology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, Boulogne-Billancourt, France
| | - Christine Longvert
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France.,Research unit EA4340 'Biomarkers and clinical trials in cancerology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, Boulogne-Billancourt, France
| | - Astrid Blom
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France.,Research unit EA4340 'Biomarkers and clinical trials in cancerology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, Boulogne-Billancourt, France
| | - Philippe Saiag
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France.,Research unit EA4340 'Biomarkers and clinical trials in cancerology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, Boulogne-Billancourt, France
| | - Elisa Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France. .,Research unit EA4340 'Biomarkers and clinical trials in cancerology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, Boulogne-Billancourt, France.
| |
Collapse
|
78
|
Abstract
Melanoma accounts for approximately 1% of all skin cancers but contributes to almost all skin cancer deaths. The developing picture suggests that melanoma phenotypes are driven by epigenetic mechanisms that reflect a complex interplay between genotype and environment. Furthermore, the growing consensus is that current classification standards, notwithstanding pertinent clinical history and appropriate biopsy, fall short of capturing the vast complexity of the disease. This article summarizes the current understanding of the clinical picture of melanoma, with a focus on the tremendous breakthroughs in molecular classification and therapeutics.
Collapse
Affiliation(s)
- Sarem Rashid
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02466, USA; Boston University School of Medicine, Boston, MA, USA
| | - Hensin Tsao
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02466, USA.
| |
Collapse
|
79
|
Wan Y, An S, Zhou Y, Zhang J, Zhang Y, Gan T, Liu Q. Is Weak Acid Beneficial for Addressing Checkpoint Inhibitor-Triggered Cancer Hyper Progression in Anti-PD1/PD-L1 Immunotherapies? Cancer Control 2021; 27:1073274820944290. [PMID: 32959668 PMCID: PMC7513399 DOI: 10.1177/1073274820944290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Numerous cases of checkpoint inhibitor–triggered cancer hyperprogression
have been documented. A previous hypothesis attributes cancer onset to
the local buildup of hydrogen chloride, jointly mediated by hydrogen
bond donors and acceptors and basic amino acids. The anti-PD1/PD-L1
immunotherapies may have caused a surge of protons or chloride ions
for the effective treatment of neoplasm, thus giving rise to the local
formation of hydrogen chloride and subsequently cancer
hyperprogression in some susceptible individuals. It was postulated
that the local strength of acidity is critical for tumor growth and
metastasis, as the intake of weak organic acids reduces cancer risks.
The anti-PD1/PD-L1 immunotherapies can be integrated with weak organic
acids to reduce adverse reactions and generate better anticancer
outcomes.
Collapse
Affiliation(s)
- Yulin Wan
- School of Life Sciences, 26469Sun Yat-sen University, Guangzhou, China
| | - Shanshan An
- School of Life Sciences, 26469Sun Yat-sen University, Guangzhou, China
| | - Yanchao Zhou
- School of Life Sciences, 26469Sun Yat-sen University, Guangzhou, China
| | - Jiaming Zhang
- School of Chemistry, 26469Sun Yat-sen University, Guangzhou, China
| | - Ying Zhang
- 214164Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Tao Gan
- School of Basic Medicine, 74554Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiuyun Liu
- School of Life Sciences, 26469Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
80
|
Dalle S, Mortier L, Corrie P, Lotem M, Board R, Arance AM, Meiss F, Terheyden P, Gutzmer R, Buysse B, Oh K, Brokaw J, Le TK, Mathias SD, Scotto J, Lord-Bessen J, Moshyk A, Kotapati S, Middleton MR. Long-term real-world experience with ipilimumab and non-ipilimumab therapies in advanced melanoma: the IMAGE study. BMC Cancer 2021; 21:642. [PMID: 34051732 PMCID: PMC8164785 DOI: 10.1186/s12885-021-08032-y] [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: 11/20/2020] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
Background Ipilimumab has shown long-term overall survival (OS) in patients with advanced melanoma in clinical trials, but robust real-world evidence is lacking. We present long-term outcomes from the IMAGE study (NCT01511913) in patients receiving ipilimumab and/or non-ipilimumab (any approved treatment other than ipilimumab) systemic therapies. Methods IMAGE was a multinational, prospective, observational study assessing adult patients with advanced melanoma treated with ipilimumab or non-ipilimumab systemic therapies between June 2012 and March 2015 with ≥3 years of follow-up. Adjusted OS curves based on multivariate Cox regression models included covariate effects. Safety and patient-reported outcomes were assessed. Results Among 1356 patients, 1094 (81%) received ipilimumab and 262 (19%) received non-ipilimumab index therapy (systemic therapy [chemotherapy, anti–programmed death 1 antibodies, or BRAF ± MEK inhibitors], radiotherapy, and radiosurgery). In the overall population, median age was 64 years, 60% were male, 78% were from Europe, and 78% had received previous treatment for advanced melanoma. In the ipilimumab-treated cohort, 780 (71%) patients did not receive subsequent therapy (IPI-noOther) and 314 (29%) received subsequent non-ipilimumab therapy (IPI-Other) on study. In the non-ipilimumab–treated cohort, 205 (78%) patients remained on or received other subsequent non-ipilimumab therapy (Other-Other) and 57 (22%) received subsequent ipilimumab therapy (Other-IPI) on study. Among 1151 patients who received ipilimumab at any time during the study (IPI-noOther, IPI-Other, and Other-IPI), 296 (26%) reported CTCAE grade ≥ 3 treatment-related adverse events, most occurring in year 1. Ipilimumab-treated and non-ipilimumab–treated patients who switched therapy (IPI-Other and Other-IPI) had longer OS than those who did not switch (IPI-noOther and Other-Other). Patients with prior therapy who did not switch therapy (IPI-noOther and Other-Other) showed similar OS. In treatment-naive patients, those in the IPI-noOther group tended to have longer OS than those in the Other-Other group. Patient-reported outcomes were similar between treatment cohorts. Conclusions With long-term follow-up (≥ 3 years), safety and OS in this real-world population of patients treated with ipilimumab 3 mg/kg were consistent with those reported in clinical trials. Patient-reported quality of life was maintained over the study period. OS analysis across both pretreated and treatment-naive patients suggested a beneficial role of ipilimumab early in treatment. Trial registration ClinicalTrials.gov, NCT01511913. Registered January 19, 2012 – Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT01511913 Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08032-y.
Collapse
Affiliation(s)
- Stéphane Dalle
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, 69495, Pierre-Bénite, France.
| | - Laurent Mortier
- Université de Lille, INSERM U1189, CHRU Lille, 59037, Lille, France
| | - Pippa Corrie
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB0 2QQ, UK
| | - Michal Lotem
- Hadassah Hebrew University Hospital, 91120, Jerusalem, Israel
| | - Ruth Board
- Royal Preston Hospital, Preston, PR2 9HT, UK
| | | | - Frank Meiss
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | | | - Ralf Gutzmer
- Medizinische Hochschule Hannover, 30625, Hanover, Germany
| | | | - Kelly Oh
- Syneos Health, Morrisville, NC, 27560, USA
| | - Jane Brokaw
- Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | - T Kim Le
- Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | | | | | | | | | | | | |
Collapse
|
81
|
Byun HK, Chang JS, Jung M, Koom WS, Chung KY, Oh BH, Roh MR, Kim KH, Lee CK, Shin SJ. Prediction of Immune-Checkpoint Blockade Monotherapy Response in Patients With Melanoma Based on Easily Accessible Clinical Indicators. Front Oncol 2021; 11:659754. [PMID: 34123816 PMCID: PMC8190329 DOI: 10.3389/fonc.2021.659754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/27/2021] [Indexed: 01/26/2023] Open
Abstract
Background Immune checkpoint blocker (ICB) has shown significant clinical activity in melanoma. However, there are no clinically approved biomarkers to aid patient selection. We aimed to identify patients with advanced or metastatic melanoma who are likely to benefit from ICB monotherapy using easily accessible clinical indicators. Materials and Methods We retrospectively reviewed the records of 134 patients with advanced or metastatic melanoma who received ICB monotherapy between 2014 and 2018. Prognostic factors of overall survival (OS) and progression-free survival (PFS) were determined using Cox regression analysis. Results During the median follow-up of 13.7 months, the median OS and PFS were 18.4 and 3.4 months, respectively. Visceral/central nervous system (CNS) metastasis (OS: adjusted hazards ratio [HR], 1.82; p=.014; PFS: HR, 1.59; p=.024), lymphopenia (<1000 cells/µL) within 3 months (OS: HR, 1.89, p=.006; PFS: HR, 1.70; p=.010), and elevated baseline lactate dehydrogenase (LDH) level (OS: HR, 2.61; p<.001; PFS: HR, 2.66; p<.001) were independent prognostic factors for both poor OS and PFS. Development of immune-related adverse events (irAE; e.g., hypothyroidism or vitiligo) within 6 months showed a trend toward better OS in multivariable analysis (HR, 0.37; p=.058). Patients with normal LDH levels and no visceral/CNS metastasis had a substantially better OS than the others (median, 40.4 vs. 13.6 months; p<.001). Among others, patients who developed irAE within 6 months achieved long-term OS (median, 43.6 vs. 13.1 months; p=.008). A decision tree was suggested using four risk factors, and the risk stratification provided significant distinction between the survival curves. Conclusion The four easily accessible clinical indicators associated with better treatment outcomes after ICB monotherapy in patients with advanced or metastatic melanoma were LDH level, the extent of disease, lymphopenia, and irAE. The combined use of these indicators can be clinically useful in improving risk stratification of patients treated with ICB monotherapy.
Collapse
Affiliation(s)
- Hwa Kyung Byun
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Jee Suk Chang
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Yang Chung
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Byung Ho Oh
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Ryung Roh
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Joon Shin
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
82
|
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: 3.0] [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
|
83
|
Vara BA, Levi SM, Achab A, Candito DA, Fradera X, Lesburg CA, Kawamura S, Lacey BM, Lim J, Methot JL, Xu Z, Xu H, Smith DM, Piesvaux JA, Miller JR, Bittinger M, Ranganath SH, Bennett DJ, DiMauro EF, Pasternak A. Discovery of Diaminopyrimidine Carboxamide HPK1 Inhibitors as Preclinical Immunotherapy Tool Compounds. ACS Med Chem Lett 2021; 12:653-661. [PMID: 33859804 PMCID: PMC8040257 DOI: 10.1021/acsmedchemlett.1c00096] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1), a serine/threonine kinase, is a negative immune regulator of T cell receptor (TCR) and B cell signaling that is primarily expressed in hematopoietic cells. Accordingly, it has been reported that HPK1 loss-of-function in HPK1 kinase-dead syngeneic mouse models shows enhanced T cell signaling and cytokine production as well as tumor growth inhibition in vivo, supporting its value as an immunotherapeutic target. Herein, we present the structurally enabled discovery of novel, potent, and selective diaminopyrimidine carboxamide HPK1 inhibitors. The key discovery of a carboxamide moiety was essential for enhanced enzyme inhibitory potency and kinome selectivity as well as sustained elevation of cellular IL-2 production across a titration range in human peripheral blood mononuclear cells. The elucidation of structure-activity relationships using various pendant amino ring systems allowed for the identification of several small molecule type-I inhibitors with promising in vitro profiles.
Collapse
Affiliation(s)
- Brandon A. Vara
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Samuel M. Levi
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Abdelghani Achab
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - David A. Candito
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Xavier Fradera
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Charles A. Lesburg
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Shuhei Kawamura
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Brian M. Lacey
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Jongwon Lim
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Joey L. Methot
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Zangwei Xu
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Haiyan Xu
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Dustin M. Smith
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Jennifer A. Piesvaux
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - J. Richard Miller
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Mark Bittinger
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Sheila H. Ranganath
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - David J. Bennett
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Erin F. DiMauro
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Alexander Pasternak
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| |
Collapse
|
84
|
Shoushtari AN. Incorporating VEGF Blockade Into a Shifting Treatment Paradigm for Mucosal Melanoma. J Clin Oncol 2021; 39:867-869. [PMID: 33492992 DOI: 10.1200/jco.20.03523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
85
|
Karimi A, Alilou S, Mirzaei HR. Adverse Events Following Administration of Anti-CTLA4 Antibody Ipilimumab. Front Oncol 2021; 11:624780. [PMID: 33767992 PMCID: PMC7985548 DOI: 10.3389/fonc.2021.624780] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/02/2021] [Indexed: 12/19/2022] Open
Abstract
Ipilimumab, a monoclonal anti-CTLA4 antibody, paved the path for promising treatments, particularly in advanced forms of numerous cancers like melanoma. By blockading CTLA-4, ipilimumab can abolish the higher binding affinity of B7 for CTLA-4, setting CD28 free to act unlimited. This blockade can result in an amplified antitumor immune response, and thereby, boosting more effective tumor regression. However, this blockage can lead to diminished self-tolerance and yielding autoimmune complications. The current review aims to describe adverse events (AEs) following the administration of ipilimumab in different cancers as every benefit comes at a cost. We will also discuss AEs in two different categories, melanoma and non-melanoma, owing to the possible shining promises in treating non-melanoma cancers. As the melanoma settings are more studied than other cancers, it might even help predict the patterns related to the other types of cancers. This similarity also might help physicians to predict adverse events and correctly manage them in non-melanoma cancers using the extensive findings reported in the more-studied melanoma settings. Recognizing the adverse events is vital since most of the adverse events could be reverted while carefully implementing guidelines. Finally, we will also describe the observed effectiveness of ipilimumab in non-melanoma cancers. This effectiveness reveals the importance of understanding the profile of adverse events in this group, even though some have not received FDA approval yet. Further clinical trials and careful systematic reviews may be required to decipher the hidden aspects of therapies with ipilimumab and its related AEs.
Collapse
Affiliation(s)
- Amirali Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanam Alilou
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
86
|
Thomas D, Bello DM. Adjuvant immunotherapy for melanoma. J Surg Oncol 2021; 123:789-797. [PMID: 33595889 DOI: 10.1002/jso.26329] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/11/2020] [Accepted: 11/23/2020] [Indexed: 02/04/2023]
Abstract
Surgical resection is the treatment for early cutaneous melanoma and is often curative. Some patients, however, will subsequently relapse. High-risk features in the primary tumor and regional lymph node metastasis highlight patient subsets that are at increased risk for recurrent disease. Immunotherapy in the form of checkpoint inhibitors ipilimumab, nivolumab, and pembrolizumab have been shown to improve recurrence-free survival for node-positive melanoma in the adjuvant setting and will be the focus of this review.
Collapse
Affiliation(s)
- Daniel Thomas
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Danielle M Bello
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
87
|
Relecom A, Merhi M, Inchakalody V, Uddin S, Rinchai D, Bedognetti D, Dermime S. Emerging dynamics pathways of response and resistance to PD-1 and CTLA-4 blockade: tackling uncertainty by confronting complexity. J Exp Clin Cancer Res 2021; 40:74. [PMID: 33602280 PMCID: PMC7893879 DOI: 10.1186/s13046-021-01872-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/08/2021] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint inhibitors provide considerable therapeutic benefit in a range of solid cancers as well as in a subgroup of hematological malignancies. Response rates are however suboptimal, and despite considerable efforts, predicting response to immune checkpoint inhibitors ahead of their administration in a given patient remains elusive. The study of the dynamics of the immune system and of the tumor under immune checkpoint blockade brought insight into the mechanisms of action of these therapeutic agents. Equally relevant are the mechanisms of adaptive resistance to immune checkpoint inhibitors that have been uncovered through this approach. In this review, we discuss the dynamics of the immune system and of the tumor under immune checkpoint blockade emanating from recent studies on animal models and humans. We will focus on mechanisms of action and of resistance conveying information predictive of therapeutic response.
Collapse
Affiliation(s)
- Allan Relecom
- Department of Medical Oncology, Translational Research Institute, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Department of Medical Oncology, Translational Research Institute, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Varghese Inchakalody
- Department of Medical Oncology, Translational Research Institute, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute & Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Darawan Rinchai
- Cancer Research Program, Research Branch, Sidra Medicine, Doha, Qatar
| | - Davide Bedognetti
- Cancer Research Program, Research Branch, Sidra Medicine, Doha, Qatar.
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy.
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| | - Said Dermime
- Department of Medical Oncology, Translational Research Institute, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar.
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
88
|
Nguyen HTM, Katta N, Widman JA, Takematsu E, Feng X, Torres-Hurtado SA, Betancourt T, Baker AB, Suggs LJ, Milner TE, Tunnell JW. Laser nanobubbles induce immunogenic cell death in breast cancer. NANOSCALE 2021; 13:3644-3653. [PMID: 33538275 PMCID: PMC8710258 DOI: 10.1039/d0nr06587k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Recent advances in immunotherapy have highlighted a need for therapeutics that initiate immunogenic cell death in tumors to stimulate the body's immune response to cancer. This study examines whether laser-generated bubbles surrounding nanoparticles ("nanobubbles") induce an immunogenic response for cancer treatment. A single nanosecond laser pulse at 1064 nm generates micron-sized bubbles surrounding gold nanorods in the cytoplasm of breast cancer cells. Cell death occurred in cells treated with nanorods and irradiated, but not in cells with irradiation treatment alone. Cells treated with nanorods and irradiation had increased damage-associated molecular patterns (DAMPs), including increased expression of chaperone proteins human high mobility group box 1 (HMGB1), adenosine triphosphate (ATP), and heat shock protein 70 (HSP70). This enhanced expression of DAMPs led to the activation of dendritic cells. Overall, this treatment approach is a rapid and highly specific method to eradicate tumor cells with simultaneous immunogenic cell death signaling, showing potential as a combination strategy for immunotherapy.
Collapse
Affiliation(s)
- Hieu T M Nguyen
- Department of Biomedical Engineering, The University of Texas at Austin, TX, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
NLRC5/CITA expression correlates with efficient response to checkpoint blockade immunotherapy. Sci Rep 2021; 11:3258. [PMID: 33547395 PMCID: PMC7865024 DOI: 10.1038/s41598-021-82729-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/21/2021] [Indexed: 12/17/2022] Open
Abstract
Checkpoint blockade-mediated immunotherapy is emerging as an effective treatment modality for multiple cancer types. However, cancer cells frequently evade the immune system, compromising the effectiveness of immunotherapy. It is crucial to develop screening methods to identify the patients who would most benefit from these therapies because of the risk of the side effects and the high cost of treatment. Here we show that expression of the MHC class I transactivator (CITA), NLRC5, is important for efficient responses to anti-CTLA-4 and anti-PD1 checkpoint blockade therapies. Melanoma tumors derived from patients responding to immunotherapy exhibited significantly higher expression of NLRC5 and MHC class I-related genes compared to non-responding patients. In addition, multivariate analysis that included the number of tumor-associated non-synonymous mutations, predicted neo-antigen load and PD-L2 expression was capable of further stratifying responders and non-responders to anti-CTLA4 therapy. Moreover, expression or methylation of NLRC5 together with total somatic mutation number were significantly correlated with increased patient survival. These results suggest that NLRC5 tumor expression, alone or together with tumor mutation load constitutes a valuable predictive biomarker for both prognosis and response to anti-CTLA-4 and potentially anti-PD1 blockade immunotherapy in melanoma patients.
Collapse
|
90
|
Botella-Estrada R, Boada-García A, Carrera-Álvarez C, Fernández-Figueras M, González-Cao M, Moreno-Ramírez D, Nagore E, Ríos-Buceta L, Rodríguez-Peralto JL, Samaniego-González E, Tejera-Vaquerizo A, Vílchez-Márquez F, Descalzo-Gallego MA, García-Doval I. Clinical Practice Guideline on Melanoma From the Spanish Academy of Dermatology and Venereology (AEDV). ACTAS DERMO-SIFILIOGRAFICAS 2021; 112:142-152. [PMID: 32721390 DOI: 10.1016/j.ad.2020.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 07/04/2020] [Indexed: 10/23/2022] Open
Abstract
Specialist approaches to the diagnosis and treatment of melanoma have undergone many changes. This guideline aims to provide Spanish dermatologists with evidence-based information for resolving the most common doubts that arise in clinical practice. Members of the Spanish Oncologic Dermatology and Surgery Group (GEDOC) with experience treating melanoma were invited to participate in drafting the guideline. The group developed a new guideline on the basis of existing ones, using the ADAPTE collaboration process, first summarizing the care process and posing relevant clinical questions, then selecting guidelines with the best scores according to the AGREE II (Appraisal of Guidelines for Research and Evaluation) tool. Finally, the group searched the selected guidelines for answers to the clinical questions, drafted recommendations, and sent them for external review. The guideline is structured around 21 clinical questions chosen for their relevance to issues that make clinical decisions about the management of melanoma difficult. Evidence from existing guidelines was used to answer the questions. A limitation of this guide derives from the scarce evidence available for answering some questions. Moreover, some areas are changing rapidly, so recommendations must be updated often. The present guideline offers answers to clinical questions about the routine management of melanoma in clinical practice and provides dermatologists with a reference to guide decisions, taking into consideration the resources available and patient preferences.
Collapse
Affiliation(s)
- R Botella-Estrada
- Servicio de Dermatología, Hospital Universitario La Fe, Valencia, España; Departamento de Medicina, Universitat de València, Valencia, España.
| | - A Boada-García
- Servicio de Dermatología, Hospital Universitario Germans Trias i Pujol, Badalona, Barcelona, España
| | | | - M Fernández-Figueras
- Área de Anatomía Patológica, Hospital Universitario General de Cataluña-Quirón Salud, Barcelona, España
| | - M González-Cao
- Translational Cancer Research Unit, Instituto Oncológico Dr. Rosell, Hospital Universitario Dexeus, Barcelona, España
| | - D Moreno-Ramírez
- Servicio de Dermatología, Hospital Universitario Virgen Macarena, Sevilla, España
| | - E Nagore
- Servicio de Dermatología, Instituto Valenciano de Oncología, Valencia, España
| | - L Ríos-Buceta
- Servicio de Dermatología, Hospital Universitario Ramón y Cajal, Madrid, España
| | - J L Rodríguez-Peralto
- Servicio de Anatomía Patológica, Hospital Universitario 12 de Octubre, Madrid, España
| | - E Samaniego-González
- Servicio de Dermatología, Complejo Asistencial Universitario de León, León, España
| | | | - F Vílchez-Márquez
- Servicio de Dermatología, Hospital Universitario Virgen de las Nieves, Granada, España
| | - M A Descalzo-Gallego
- Unidad de Investigación, Fundación Piel Sana, Academia Española de Dermatología y Venereología, Madrid, España
| | - I García-Doval
- Unidad de Investigación, Fundación Piel Sana, Academia Española de Dermatología y Venereología, Madrid, España
| |
Collapse
|
91
|
Gault A, Anderson A, Plummer R, Stewart C, Pratt A, Rajan N. Cutaneous immune‐related adverse events in patients with melanoma treated with checkpoint inhibitors. Br J Dermatol 2021; 185:263-271. [DOI: 10.1111/bjd.19750] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2020] [Indexed: 12/24/2022]
Affiliation(s)
- A. Gault
- Translational and Clinical Research Institute Newcastle University Newcastle upon Tyne UK
- Northern Centre for Cancer Care Freeman Hospital Newcastle Upon Tyne UK
| | - A.E. Anderson
- Translational and Clinical Research Institute Newcastle University Newcastle upon Tyne UK
| | - R. Plummer
- Translational and Clinical Research Institute Newcastle University Newcastle upon Tyne UK
- Northern Centre for Cancer Care Freeman Hospital Newcastle Upon Tyne UK
| | - C. Stewart
- Translational and Clinical Research Institute Newcastle University Newcastle upon Tyne UK
| | - A.G. Pratt
- Translational and Clinical Research Institute Newcastle University Newcastle upon Tyne UK
| | - N. Rajan
- Translational and Clinical Research Institute Newcastle University Newcastle upon Tyne UK
- Department of Dermatology Royal Victoria Infirmary Newcastle upon Tyne UK
| |
Collapse
|
92
|
Botella-Estrada R, Boada-García A, Carrera-Álvarez C, Fernández-Figueras M, González-Cao M, Moreno-Ramírez D, Nagore E, Ríos-Buceta L, Rodríguez-Peralto J, Samaniego-González E, Tejera-Vaquerizo A, Vílchez-Márquez F, Descalzo-Gallego M, García-Doval I. Clinical Practice Guideline on Melanoma From the Spanish Academy of Dermatology and Venereology (AEDV). ACTAS DERMO-SIFILIOGRAFICAS 2021. [DOI: 10.1016/j.adengl.2020.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
93
|
Al-Adra DP, Hammel L, Roberts J, Woodle ES, Levine D, Mandelbrot D, Verna E, Locke J, D'Cunha J, Farr M, Sawinski D, Agarwal PK, Plichta J, Pruthi S, Farr D, Carvajal R, Walker J, Zwald F, Habermann T, Gertz M, Bierman P, Dizon DS, Langstraat C, Al-Qaoud T, Eggener S, Richgels JP, Chang GJ, Geltzeiler C, Sapisochin G, Ricciardi R, Krupnick AS, Kennedy C, Mohindra N, Foley DP, Watt KD. Preexisting melanoma and hematological malignancies, prognosis, and timing to solid organ transplantation: A consensus expert opinion statement. Am J Transplant 2021; 21:475-483. [PMID: 32976703 PMCID: PMC8555431 DOI: 10.1111/ajt.16324] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 01/25/2023]
Abstract
Patients undergoing evaluation for solid organ transplantation (SOT) frequently have a history of malignancy. Only patients with treated cancer are considered for SOT but the benefits of transplantation need to be balanced against the risk of tumor recurrence, taking into consideration the potential effects of immunosuppression. Prior guidelines on timing to transplant in patients with a prior treated malignancy do not account for current staging, disease biology, or advances in cancer treatments. To update these recommendations, the American Society of Transplantation (AST) facilitated a consensus workshop to comprehensively review contemporary literature regarding cancer therapies, cancer stage specific prognosis, the kinetics of cancer recurrence, as well as the limited data on the effects of immunosuppression on cancer-specific outcomes. This document contains prognosis, treatment, and transplant recommendations for melanoma and hematological malignancies. Given the limited data regarding the risk of cancer recurrence in transplant recipients, the goal of the AST-sponsored conference and the consensus documents produced are to provide expert opinion recommendations that help in the evaluation of patients with a history of a pretransplant malignancy for transplant candidacy.
Collapse
Affiliation(s)
- David P Al-Adra
- Department of Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin
| | - Laura Hammel
- Department of Anesthesiology, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin
| | - John Roberts
- Department of Surgery, University of California San Francisco, San Francisco, California
| | - E Steve Woodle
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Deborah Levine
- Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Didier Mandelbrot
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin
| | - Elizabeth Verna
- Department of Medicine, New York-Presbyterian/Columbia, New York, New York
| | - Jayme Locke
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Maryjane Farr
- Department of Medicine, New York-Presbyterian/Columbia, New York, New York
| | - Deirdre Sawinski
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Jennifer Plichta
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Sandhya Pruthi
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Deborah Farr
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Richard Carvajal
- Department of Medicine, New York-Presbyterian/Columbia, New York, New York
| | - John Walker
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Fiona Zwald
- Piedmont Transplant Institute, Piedmont Atlanta Hospital, Atlanta, Georgia
| | | | - Morie Gertz
- Hematology Division, Mayo Clinic, Rochester, Minnesota, USA
| | - Philip Bierman
- Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Don S Dizon
- Lifespan Cancer Institute and Brown University, Providence, Rhode Island
| | - Carrie Langstraat
- Departments of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Talal Al-Qaoud
- Department of Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin
| | - Scott Eggener
- Department of Urology, University of Chicago, Chicago, Illinois
| | - John P Richgels
- Department of Urology, University of Chicago, Chicago, Illinois
| | - George J Chang
- Department of Surgical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Cristina Geltzeiler
- Department of Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin
| | | | - Rocco Ricciardi
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Cassie Kennedy
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Nisha Mohindra
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - David P Foley
- Department of Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin
| | | |
Collapse
|
94
|
Wu J, Das J, Kalra M, Ratto B. Comparative efficacy of dabrafenib + trametinib versus treatment options for metastatic melanoma in first-line settings. J Comp Eff Res 2021; 10:267-280. [PMID: 33448878 DOI: 10.2217/cer-2020-0249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: The objective was to systematically review the literature and assess the relative efficacy of agents approved in first-line settings via network meta-analysis. Materials & methods: A literature review was conducted via searching different medical databases. The eligibility criteria included Phase II or III randomized controlled trials that had enrolled treatment-naive adult patients with advanced/metastatic melanoma. Results: The network meta-analysis results suggested that dabrafenib + trametinib significantly prolongs the survival outcomes compared with the monotherapies and had comparable efficacy profile compared with encorafenib + binimetinib and cobimetinib + vemurafenib. In comparison with immunotherapies, the results varied for progression-free survival and overall survival. Conclusion: Long-term survival data of dabrafenib + trametinib establishes the combination as one of the preferred treatment options for previously untreated melanoma patients.
Collapse
Affiliation(s)
- Jing Wu
- School of Pharmaceutical Science & Technology, Tianjin University, PR China
| | - Jaydeep Das
- Novartis Healthcare Pvt Ltd, Hyderabad, India
| | - Manik Kalra
- Novartis Healthcare Pvt Ltd, Hyderabad, India
| | - Barbara Ratto
- Novartis Pharmaceuticals Corporation, NJ 07936-1080, US
| |
Collapse
|
95
|
Sood S, Jayachandiran R, Pandey S. Current Advancements and Novel Strategies in the Treatment of Metastatic Melanoma. Integr Cancer Ther 2021; 20:1534735421990078. [PMID: 33719631 PMCID: PMC8743966 DOI: 10.1177/1534735421990078] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/07/2020] [Accepted: 01/06/2021] [Indexed: 02/03/2023] Open
Abstract
Melanoma is the deadliest form of skin cancer in the world with a growing incidence in North America. Contemporary treatments for melanoma include surgical resection, chemotherapy, and radiotherapy. However, apart from resection in early melanoma, the prognosis of patients using these treatments is typically poor. In the past decade, there have been significant advancements in melanoma therapies. Immunotherapies such as ipilimumab and targeted therapies such as vemurafenib have emerged as a promising option for patients as seen in both scientific and clinical research. Furthermore, combination therapies are starting to be administered in the form of polychemotherapy, polyimmunotherapy, and biochemotherapy, of which some have shown promising outcomes in relative efficacy and safety due to their multiple targets. Alongside these treatments, new research has been conducted into the evidence-based use of natural health products (NHPs) and natural compounds (NCs) on melanoma which may provide a long-term and non-toxic form of complementary therapy. Nevertheless, there is a limited consolidation of the research conducted in emerging melanoma treatments which may be useful for researchers and clinicians. Thus, this review attempts to evaluate the therapeutic efficacy of current advancements in metastatic melanoma treatment by surveying new research into the molecular and cellular basis of treatments along with their clinical efficacy. In addition, this review aims to elucidate novel strategies that are currently being used and have the potential to be used in the future.
Collapse
|
96
|
Holmberg CJ, Katsarelias D, Jespersen H, Carneiro A, Elander NO, Helgadottir H, Isaksson K, Jansson M, Wirén S, Ullenhag GJ, Ny L, Olofsson Bagge R. Surgery of metastatic melanoma after systemic therapy - the SUMMIST trial: study protocol for a randomized controlled trial. Acta Oncol 2021; 60:52-55. [PMID: 33252282 DOI: 10.1080/0284186x.2020.1846213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Carl Jacob Holmberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Dimitrios Katsarelias
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Jespersen
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Akershus University Hospital, Nordbyhagen, Norway
| | - Ana Carneiro
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Nils O. Elander
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hildur Helgadottir
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Karolin Isaksson
- Department of Clinical Sciences, Surgery, Lund University, Lund, Sweden
- Department of Surgery, Central Hospital Kristianstad, Kristianstad, Sweden
| | - Malin Jansson
- Department of Surgical and Perioperative Sciences, Umeå University and Umeå University Hospital, Umeå, Sweden
| | - Sara Wirén
- Department of Radiation Sciences, Oncology, Umeå University Hospital, Umeå, Sweden
| | - Gustav J. Ullenhag
- Department of Immunology, Genetics and Pathology, Science of Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
97
|
Cybulska-Stopa B, Pacholczak-Madej R, Kamińska-Winciorek G, Ziętek M, Czarnecka AM, Piejko K, Galus Ł, Ziółkowska B, Kieszko S, Kempa-Kamińska N, Calik J, Rolski J, Sałek-Zań A, Gajewska-Wicher K, Drosik-Kwaśniewska A, Rogala P, Kubiatowski T, Suwiński R, Mackiewicz J, Rutkowski P. First-line treatment of advanced/metastatic melanoma with anti-PD-1 antibodies: multicenter experience in Poland. Immunotherapy 2020; 13:297-307. [PMID: 33353420 DOI: 10.2217/imt-2020-0217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: To evaluate treatment results in advanced/metastatic melanoma patients treated with anti-PD-1 immunotherapy in routine practice in oncology centers in Poland. Methods: Multicenter retrospective analysis included 499 patients with unresectable/metastatic (stage IIIC-IV) melanoma treated with anti-PD-1 in first-line therapy. Results: Estimated median overall survival (OS) and progression-free survival (PFS) were 19.9 and 7.9 months, respectively. Multivariate analysis confirmed that ECOG 0, no brain metastases, normal lactate dehydrogenase level and occurrence of immune-related adverse events (irAEs) were statistically significantly associated with improved OS and PFS. Any irAE occurred in 24% of patients. Grade 3 or Grade 4 irAEs occurred in 6% of patients. Conclusion: Analysis revealed a slightly worse OS in real-world treatment in comparison to clinical trials (KEYNOTE-006 and CheckMate 066). Polish population treatment results are similar to other studies of real-world data. PFS and ORR are similar in our research and clinical trials.
Collapse
Affiliation(s)
- Bożena Cybulska-Stopa
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Krakow, Poland
| | - Renata Pacholczak-Madej
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Krakow, Poland.,Department of Anatomy, Jagiellonian University, Medical College, 31-543 Krakow, Poland
| | - Grażyna Kamińska-Winciorek
- Department of Bone Marrow Transplantation & Hematology-Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Marcin Ziętek
- Department of Oncological Surgery - Skin Cancer Unit, Lower Silesian Oncology Center, 53-413 Wroclaw, Poland.,Department of Oncological Surgery, Wroclaw Medical University, 53-413 Wroclaw, Poland
| | - Anna M Czarnecka
- Department of Soft Tissue/Bone Sarcoma & Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland.,Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Karolina Piejko
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Krakow, Poland
| | - Łukasz Galus
- Department of Medical & Experimental Oncology, Poznan University of Medical Sciences, 60-786 Poznan, Poland.,Chemotherapy Department, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Barbara Ziółkowska
- II Clinic of Radiotherapy & Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Stanisław Kieszko
- Department of Clinical Oncology, Saint Jan of Dukla Oncology Centre of the Lublin Region, 20-090 Lublin, Poland
| | | | - Jacek Calik
- Department of Clinical Oncology, Lower Silesian Oncology Center, 53-413 Wroclaw, Poland
| | - Janusz Rolski
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Krakow, Poland
| | - Agata Sałek-Zań
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Krakow, Poland
| | - Katarzyna Gajewska-Wicher
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Krakow, Poland
| | - Anna Drosik-Kwaśniewska
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Krakow, Poland
| | - Paweł Rogala
- Department of Soft Tissue/Bone Sarcoma & Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Tomasz Kubiatowski
- Department of Clinical Oncology, Saint Jan of Dukla Oncology Centre of the Lublin Region, 20-090 Lublin, Poland
| | - Rafał Suwiński
- II Clinic of Radiotherapy & Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Jacek Mackiewicz
- Department of Medical & Experimental Oncology, Poznan University of Medical Sciences, 60-786 Poznan, Poland.,Department of Diagnostics & Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma & Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
98
|
Deng W, Wang Y, Liu X, Liu J, Wang L, Yang Z, Yang M, An Y, Tang C, Sanford NN, Kim BYS, Jiang W. Assessment of Trends in Second Primary Cancers in Patients With Metastatic Melanoma From 2005 to 2016. JAMA Netw Open 2020; 3:e2028627. [PMID: 33295975 PMCID: PMC7726633 DOI: 10.1001/jamanetworkopen.2020.28627] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
IMPORTANCE To date, the risk of developing second primary cancers (SPCs) after the first primary melanoma has not been studied in the era of immune checkpoint inhibitors (ICIs). OBJECTIVE To assess differences in the risk of SPCs in patients with primary melanoma before (2005-2010) and after (2011-2016) the introduction and approval of ICIs. DESIGN, SETTING, AND PARTICIPANTS Population-based cohort study using the Surveillance, Epidemiology, and End Results database from January 2005 to December 2016 of patients diagnosed with metastatic melanoma. Data were analyzed from January 4 to June 30, 2020. EXPOSURES Receipt of immunotherapy or other anticancer agents. MAIN OUTCOMES AND MEASURES The primary outcome was the development of second primary cancers in patients with melanoma. Standardized incidence ratios (SIRs) were calculated for the development of SPCs before and after the introduction of ICIs. RESULTS Among 5016 patients with diagnosed metastatic melanoma, 2888 (58%) were younger than 65 years at the time of diagnosis, and 3441 (69%) were male. From 2005 to 2010, SIRs were 3.24 (95% CI, 0.08-18.04) for small intestine cancer, 1.93 (95% CI, 1.14-3.05) for lung and bronchus cancer, 2.77 (95% CI, 1.02-6.03) for kidney cancer, and 7.29 (95% CI, 2.93-15.02) for myeloma. From 2011 to 2016, SIRs were 9.23 (95% CI, 1.12-33.35) for small intestine cancer, 1.54 (95% CI, 0.71-2.93) for lung and bronchus cancer, 2.66 (95% CI, 0.73-6.82) for kidney cancer, and 5.90 (95% CI, 1.61-15.10) for myeloma. The overall risk of developing SPCs in individuals who survived the first primary melanoma was 65% higher (SIR, 1.65; 95% CI, 1.35-2.00) in the pre-ICIs period and 98% higher (SIR, 1.98; 95% CI, 1.57-2.45) in the post-ICIs period than the overall cancer incidence rate in the general population. CONCLUSIONS AND RELEVANCE In this study, an increase in the overall risk of second primary cancers after melanoma after the introduction of immune checkpoint inhibitors was observed. The pattern of SPCs has been altered in the era of systemic therapy. Close monitoring and screening for SPCs may be warranted in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Weiye Deng
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas
| | - Yifan Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas
| | - Xiangyu Liu
- Department of Biostatistics and Data Science, School of Public Health, the University of Texas Health Science Center, Houston
| | - Jieqiong Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Wang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Zhaogang Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas
| | - Mingming Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas
| | - Yi An
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| | - Chad Tang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Nina N. Sanford
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas
| | - Betty Y. S. Kim
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston
| | - Wen Jiang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
99
|
Farrow NE, Turner MC, Salama AKS, Beasley GM. Overall Survival Improved for Contemporary Patients with Melanoma: A 2004-2015 National Cancer Database Analysis. Oncol Ther 2020; 8:261-275. [PMID: 32700043 PMCID: PMC7683754 DOI: 10.1007/s40487-020-00117-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Since 2011, encouraging clinical trial results have led to approval of multiple new therapies for advanced melanoma, but the impact of these therapies outside of trial populations is largely unknown. This study examines use of novel therapies and survival in contemporary patients with melanoma. METHODS Stage I-IV melanoma patients were identified in the 2004-2015 National Cancer Database and grouped into historic (2004-2010) and contemporary (2011-2015) cohorts. Overall survival (OS) was compared using Kaplan-Meier and Cox proportional hazard modeling adjusting for patient, tumor, and facility characteristics. RESULTS Of 268,668 patients, 136,828 were classified as historic and 131,840 as contemporary. Among all stages, immunotherapy utilization was significantly higher among contemporary patients (5.3% vs. 5.1%, p = 0.006). Adjusted OS was improved in the contemporary cohort (hazard ratio [HR]: 0.90 p < 0.001). There was no difference in OS among stage I/II patients between groups (HR: 0.99, p = 0.63), while OS was significantly improved for contemporary stage III/IV patients (HR: 0.85, p < 0.001). Among stage III/IV patients who received immunotherapy, OS was improved for the contemporary cohort (HR: 0.87, p = 0.014). CONCLUSIONS Adjusted overall survival for contemporary melanoma patients is improved. This effect is driven by improvements for those with advanced stage disease, particularly those that received immunotherapy and BRAF/MEK targeted therapies.
Collapse
Affiliation(s)
- Norma E Farrow
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Megan C Turner
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - April K S Salama
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
100
|
Neek M, Tucker JA, Butkovich N, Nelson EL, Wang SW. An Antigen-Delivery Protein Nanoparticle Combined with Anti-PD-1 Checkpoint Inhibitor Has Curative Efficacy in an Aggressive Melanoma Model. ADVANCED THERAPEUTICS 2020; 3:2000122. [PMID: 34141865 PMCID: PMC8205422 DOI: 10.1002/adtp.202000122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibition is a promising alternative treatment to standard chemotherapies; however, it fails to achieve long-term remission in a significant portion of patients. A previously developed protein nanoparticle-based platform (E2 nanoparticle) delivers cancer antigens to increase antigen-specific tumor responses. While prior work has focussed on prophylactic conditions, the objectives in this study are therapeutic. It is hypothesized that immune checkpoint inhibition, when augmented by antigen delivery using E2 nanoparticles containing CpG oligonucleotide 1826 (CpG) and a glycoprotein 100 (gp100) melanoma antigen epitope (CpG-gp-E2), would synergistically elicit antitumor responses. To identify a regimen primed for obtaining effective treatment results, immune benchmarks in the spleen and tumor are examined. Conditions that lead to significant immune activation, including increases in gp100-specific interferon gamma (IFN-𝜸), CD8 T cells in the spleen, tumor-infiltrating CD8 T cells, and survival time are identified. Based on the findings, the resulting combination of CpG-gp-E2 and anti-programmed cell death protein 1 (anti-PD-1) treatment in tumor-challenged mice yield significantly increased long-term survival; more than 50% of the mice treated with combination therapy were tumor-free, compared with 0% and ≈5% for CpG-gp-E2 and anti-PD-1 alone, respectively. Evidence of a durable antitumor response is also observed upon tumor rechallenge, pointing to long-lasting immune memory.
Collapse
Affiliation(s)
- Medea Neek
- Department of Chemical and Biomolecular Engineering University of California Irvine, CA 92697, USA
| | - Jo Anne Tucker
- Department of Medicine University of California Irvine, CA 92697, USA
| | - Nina Butkovich
- Department of Chemical and Biomolecular Engineering University of California Irvine, CA 92697, USA
| | - Edward L Nelson
- Department of Medicine University of California Irvine, CA 92697, USA
| | - Szu-Wen Wang
- Department of Chemical and Biomolecular Engineering University of California Irvine, CA 92697, USA
| |
Collapse
|