1
|
Butterfield LH, Najjar YG. Immunotherapy combination approaches: mechanisms, biomarkers and clinical observations. Nat Rev Immunol 2024; 24:399-416. [PMID: 38057451 PMCID: PMC11460566 DOI: 10.1038/s41577-023-00973-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
The approval of the first immune checkpoint inhibitors provided a paradigm shift for the treatment of malignancies across a broad range of indications. Whereas initially, single-agent immune checkpoint inhibition was used, increasing numbers of patients are now treated with combination immune checkpoint blockade, where non-redundant mechanisms of action of the individual agents generally lead to higher response rates. Furthermore, immune checkpoint therapy has been combined with various other therapeutic modalities, including chemotherapy, radiotherapy and other immunotherapeutics such as vaccines, adoptive cellular therapies, cytokines and others, in an effort to maximize clinical efficacy. Currently, a large number of clinical trials test combination therapies with an immune checkpoint inhibitor as a backbone. However, proceeding without inclusion of broad, if initially exploratory, biomarker investigations may ultimately slow progress, as so far, few combinations have yielded clinical successes based on clinical data alone. Here, we present the rationale for combination therapies and discuss clinical data from clinical trials across the immuno-oncology spectrum. Moreover, we discuss the evolution of biomarker approaches and highlight the potential new directions that comprehensive biomarker studies can yield.
Collapse
Affiliation(s)
- Lisa H Butterfield
- University of California San Francisco, Microbiology and Immunology, San Francisco, CA, USA.
| | | |
Collapse
|
2
|
Mo Z, Liu J, Zhang J, Deng Y, Xu M, Jiang Y. Association of NRAS mutations and tertiary lymphoid structure formation with clinical outcomes of adjuvant PD-1 inhibitors for acral melanoma. Int Immunopharmacol 2023; 124:110973. [PMID: 37769536 DOI: 10.1016/j.intimp.2023.110973] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVES This study evaluates the efficacy of programmed death-1 (PD-1) inhibitors as adjuvant therapy for acral melanoma (AM) and the predictive value of genetic mutations and tertiary lymphoid structures (TLSs). METHODS AND RESULTS A single-center retrospective longitudinal cohort study was conducted between October 1, 2018, and September 31, 2022. Patients with stages II-III completely resected AM were treated with at least two doses of adjuvant PD-1 inhibitors. A total of 44 participants were included in the final analysis, of which 41 patients with stage III. The median follow-up time, median relapse-free survival (RFS), and median distance metastasis-free survival (DMFS) for all patients were 18.4 months, 21.6 months, and 30.6 months, respectively. 21 (47.7%) and 20 (45.5%) patients were intravenously administered pembrolizumab and toripalimab, respectively. There were no significant differences in RFS (24.4 months vs. 18.9 months, p = 0.432) or DMFS (30.6 months vs. not reached, p = 0.865) between the pembrolizumab and toripalimab groups, respectively. The median DMFS (41.1 months vs. 9.0 months, p < 0.001) in the wild-type NRAS group was significantly longer than that in the NRAS mutation group. Overall, different levels of TLSs infiltration did not significantly affect patient survival. Only three people discontinued treatment due to adverse events. No treatment-related death occurred during the study period. CONCLUSION Our study suggests that adjuvant toripalimab and pembrolizumab therapy have comparable efficacies in patients with AM and are both well tolerated. Adjuvant monotherapy with PD-1 inhibitors may not be appropriate for AM with NRAS mutations.
Collapse
Affiliation(s)
- Zeming Mo
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Liu
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jinyan Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yaotiao Deng
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Miao Xu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China.
| | - Yu Jiang
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Haist M, Stege H, Kuske M, Bauer J, Klumpp A, Grabbe S, Bros M. Combination of immune-checkpoint inhibitors and targeted therapies for melanoma therapy: The more, the better? Cancer Metastasis Rev 2023; 42:481-505. [PMID: 37022618 PMCID: PMC10348973 DOI: 10.1007/s10555-023-10097-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 04/07/2023]
Abstract
The approval of immune-checkpoint inhibitors (CPI) and mitogen activated protein kinase inhibitors (MAPKi) in recent years significantly improved the treatment management and survival of patients with advanced malignant melanoma. CPI aim to counter-act receptor-mediated inhibitory effects of tumor cells and immunomodulatory cell types on effector T cells, whereas MAPKi are intended to inhibit tumor cell survival. In agreement with these complementary modes of action preclinical data indicated that the combined application of CPI and MAPKi or their optimal sequencing might provide additional clinical benefit. In this review the rationale and preclinical evidence that support the combined application of MAPKi and CPI either in concurrent or consecutive regimens are presented. Further, we will discuss the results from clinical trials investigating the sequential or combined application of MAPKi and CPI for advanced melanoma patients and their implications for clinical practice. Finally, we outline mechanisms of MAPKi and CPI cross-resistance which limit the efficacy of currently available treatments, as well as combination regimens.
Collapse
Affiliation(s)
- Maximilian Haist
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Michael Kuske
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Julia Bauer
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Annika Klumpp
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
4
|
Sosa Cuevas E, Saas P, Aspord C. Dendritic Cell Subsets in Melanoma: Pathophysiology, Clinical Prognosis and Therapeutic Exploitation. Cancers (Basel) 2023; 15:cancers15082206. [PMID: 37190135 DOI: 10.3390/cancers15082206] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Evasion from immunity is a hallmark of cancer development. Dendritic cells (DCs) are strategic immune cells shaping anti-tumor immune responses, but tumor cells exploit DC versatility to subvert their functions. Unveiling the puzzling role of DCs in the control of tumor development and mechanisms of tumor-induced DC hijacking is critical to optimize current therapies and to design future efficient immunotherapies for melanoma. Dendritic cells, crucially positioned at the center of anti-tumor immunity, represent attractive targets to develop new therapeutic approaches. Harnessing the potencies of each DC subset to trigger appropriate immune responses while avoiding their subversion is a challenging yet promising step to achieve tumor immune control. This review focuses on advances regarding the diversity of DC subsets, their pathophysiology and impact on clinical outcome in melanoma patients. We provide insights into the regulation mechanisms of DCs by the tumor, and overview DC-based therapeutic developments for melanoma. Further insights into DCs' diversity, features, networking, regulation and shaping by the tumor microenvironment will allow designing novel effective cancer therapies. The DCs deserve to be positioned in the current melanoma immunotherapeutic landscape. Recent discoveries strongly motivate exploitation of the exceptional potential of DCs to drive robust anti-tumor immunity, offering promising tracks for clinical successes.
Collapse
Affiliation(s)
- Eleonora Sosa Cuevas
- EFS AuRA, R&D Laboratory, 38000 Grenoble, France
- Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Université Grenoble Alpes, 38000 Grenoble, France
| | - Philippe Saas
- EFS AuRA, R&D Laboratory, 38000 Grenoble, France
- Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Université Grenoble Alpes, 38000 Grenoble, France
| | - Caroline Aspord
- EFS AuRA, R&D Laboratory, 38000 Grenoble, France
- Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Université Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
5
|
Huang B, Su W, Yu D. Data-driven analysis to identify prognostic immune-related biomarkers in BRAF mutated cutaneous melanoma microenvironment. Front Genet 2022; 13:1081418. [DOI: 10.3389/fgene.2022.1081418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022] Open
Abstract
Skin cutaneous melanoma is one of the deadly diseases, and more than 50% of the patients have BRAF gene mutations. Evidence suggests that oncogenic BRAF modulates the immune system’s ability to recognize SKCM cells. Due to the complexity of the tumor microenvironment (TME) and a lack of a rational mechanistic basis, it is urgent to investigate the immune infiltration and identify prognostic biomarkers in BRAF mutated SKCM patients. Multiple methods including ESTIMATE algorithm, differential gene analysis, prognostic analysis and immune infiltration analysis were performed to investigate the tumor microenvironment. Based on the patient’s immune score and stromal score, immune-related genes DEGs were identified. Functional analysis revealed that these genes were mainly enriched in biological processes such as immune response, defense response and positive regulation of immune system. Furthermore, we analyzed the immune infiltrating cell components of BRAF mutated patients and revealed 4 hub genes associated with overall survival time. Several cells (Monocyte, Macrophage and Gamma delta cells) have been found to be significantly decreased in immune-high BRAF mutated SKCM group. While CD4+T, CD8+T, CD4 naïve, Tr1, Th2 and many T cell subsets were significantly increased in immune-high group. These immune cells and genes were closely related to each other. This study revealed that the dysregulation of immune function and immune cells may contribute to the poor outcomes of BRAF mutated patients. It is of great significance to our further understanding of the TME and immune dysfunction in BRAF mutated SKCM.
Collapse
|
6
|
Caksa S, Baqai U, Aplin AE. The future of targeted kinase inhibitors in melanoma. Pharmacol Ther 2022; 239:108200. [PMID: 35513054 PMCID: PMC10187889 DOI: 10.1016/j.pharmthera.2022.108200] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022]
Abstract
Melanoma is a cancer of the pigment-producing cells of the body and its incidence is rising. Targeted inhibitors that act against kinases in the MAPK pathway are approved for BRAF-mutant metastatic cutaneous melanoma and increase patients' survival. Response to these therapies is limited by drug resistance and is less durable than with immune checkpoint inhibition. Conversely, rare melanoma subtypes have few therapeutic options for advanced disease and MAPK pathway targeting agents show minimal anti-tumor effects. Nevertheless, there is a future for targeted kinase inhibitors in melanoma: in new applications such as adjuvant or neoadjuvant therapy and in novel combinations with immunotherapies or other targeted therapies. Pre-clinical studies continue to identify tumor dependencies and their corresponding actionable drug targets, paving the way for rational targeted kinase inhibitor combinations as a personalized medicine approach for melanoma.
Collapse
Affiliation(s)
- Signe Caksa
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Usman Baqai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
7
|
Avery TY, Köhler N, Zeiser R, Brummer T, Ruess DA. Onco-immunomodulatory properties of pharmacological interference with RAS-RAF-MEK-ERK pathway hyperactivation. Front Oncol 2022; 12:931774. [PMID: 35965494 PMCID: PMC9363660 DOI: 10.3389/fonc.2022.931774] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/30/2022] [Indexed: 12/25/2022] Open
Abstract
Hyperactivation of the RAS-RAF-MEK-ERK cascade - a mitogen-activated protein kinase pathway – has a well-known association with oncogenesis of leading tumor entities, including non-small cell lung cancer, colorectal carcinoma, pancreatic ductal adenocarcinoma, and malignant melanoma. Increasing evidence shows that genetic alterations leading to RAS-RAF-MEK-ERK pathway hyperactivation mediate contact- and soluble-dependent crosstalk between tumor, tumor microenvironment (TME) and the immune system resulting in immune escape mechanisms and establishment of a tumor-sustaining environment. Consequently, pharmacological interruption of this pathway not only leads to tumor-cell intrinsic disruptive effects but also modification of the TME and anti-tumor immunomodulation. At the same time, the importance of ERK signaling in immune cell physiology and potentiation of anti-tumor immune responses through ERK signaling inhibition within immune cell subsets has received growing appreciation. Specifically, a strong case was made for targeted MEK inhibition due to promising associated immune cell intrinsic modulatory effects. However, the successful transition of therapeutic agents interrupting RAS-RAF-MEK-ERK hyperactivation is still being hampered by significant limitations regarding durable efficacy, therapy resistance and toxicity. We here collate and summarize the multifaceted role of RAS-RAF-MEK-ERK signaling in physiology and oncoimmunology and outline the rationale and concepts for exploitation of immunomodulatory properties of RAS-RAF-MEK-ERK inhibition while accentuating the role of MEK inhibition in combinatorial and intermittent anticancer therapy. Furthermore, we point out the extensive scientific efforts dedicated to overcoming the challenges encountered during the clinical transition of various therapeutic agents in the search for the most effective and safe patient- and tumor-tailored treatment approach.
Collapse
Affiliation(s)
- Thomas Yul Avery
- Department of General and Visceral Surgery, Center of Surgery, Medical Center University of Freiburg, Freiburg, Germany
- *Correspondence: Thomas Yul Avery, ; Dietrich Alexander Ruess,
| | - Natalie Köhler
- Department of Medicine I - Medical Center, Medical Center University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I - Medical Center, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Tilman Brummer
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Faculty of Medicine, Medical Center University of Freiburg, Freiburg, Germany
| | - Dietrich Alexander Ruess
- Department of General and Visceral Surgery, Center of Surgery, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- *Correspondence: Thomas Yul Avery, ; Dietrich Alexander Ruess,
| |
Collapse
|
8
|
Zhang Z, Richmond A, Yan C. Immunomodulatory Properties of PI3K/AKT/mTOR and MAPK/MEK/ERK Inhibition Augment Response to Immune Checkpoint Blockade in Melanoma and Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms23137353. [PMID: 35806358 PMCID: PMC9266842 DOI: 10.3390/ijms23137353] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 02/03/2023] Open
Abstract
Hyperactivation of PI3K/AKT/mTOR and MAPK/MEK/ERK signaling pathways is commonly observed in many cancers, including triple-negative breast cancer (TNBC) and melanoma. Moreover, the compensatory upregulation of the MAPK/MEK/ERK pathway has been associated with therapeutic resistance to targeted inhibition of the PI3K/AKT/mTOR pathway, and vice versa. The immune-modulatory effects of both PI3K and MAPK inhibition suggest that inhibition of these pathways might enhance response to immune checkpoint inhibitors (ICIs). ICIs have become the standard-of-care for metastatic melanoma and are recently an option for TNBC when combined with chemotherapy, but alternative options are needed when resistance develops. In this review, we present the current mechanistic understandings, along with preclinical and clinical evidence, that outline the efficacy and safety profile of combinatorial or sequential treatments with PI3K inhibitors, MAPK inhibitors, and ICIs for treatment of malignant melanoma and metastatic TNBC. This approach may present a potential strategy to overcome resistance in patients who are a candidate for ICI therapy with tumors harboring either or both of these pathway-associated mutations.
Collapse
Affiliation(s)
- Zhizhu Zhang
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; (Z.Z.); (A.R.)
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; (Z.Z.); (A.R.)
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Chi Yan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; (Z.Z.); (A.R.)
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
- Correspondence:
| |
Collapse
|
9
|
Callahan MK, Chapman PB. PD-1 or PD-L1 Blockade Adds Little to Combination of BRAF and MEK Inhibition in the Treatment of BRAF V600-Mutated Melanoma. J Clin Oncol 2022; 40:1393-1395. [PMID: 35030031 PMCID: PMC9061142 DOI: 10.1200/jco.21.02801] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 11/20/2022] Open
|
10
|
Bari S, Muzaffar J, Eroglu Z. Combination targeted and immune therapy in the treatment of advanced melanoma: a valid treatment option for patients? Ther Adv Med Oncol 2022; 14:17588359221090306. [PMID: 35478991 PMCID: PMC9036333 DOI: 10.1177/17588359221090306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Melanomas harboring an activating BRAFV600 mutation account for 50% of all advanced melanomas. The approval of BRAF-targeted therapy revolutionized treatment of these patients with achievement of impressive responses. However, development of resistance to these drugs is a significant problem, and as such, duration of response remains low, with median progression free survival of around 11–15 months. Immune checkpoint blockers exploit the immune system to eradicate cancer and can produce durable disease control that results in long-term, treatment-free survival in some patients. These drugs have shown very impressive survival in patients with BRAF-mutated melanoma. Thus, there is a need to continue to utilize emerging data to achieve long-term disease control for patients with advanced melanoma. Combining targeted therapy with immune therapy may be one possible way to achieve this goal. In this review, the mechanisms of action of these two pathways, including the mechanistic basis of this combination, are summarized, along with results of completed and ongoing trials in triple therapy.
Collapse
Affiliation(s)
- Shahla Bari
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jameel Muzaffar
- Department of Head and Neck Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
11
|
Raftopoulou S, Valadez-Cosmes P, Mihalic ZN, Schicho R, Kargl J. Tumor-Mediated Neutrophil Polarization and Therapeutic Implications. Int J Mol Sci 2022; 23:3218. [PMID: 35328639 PMCID: PMC8951452 DOI: 10.3390/ijms23063218] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/25/2022] [Accepted: 03/09/2022] [Indexed: 01/04/2023] Open
Abstract
Neutrophils are immune cells with reported phenotypic and functional plasticity. Tumor-associated neutrophils display many roles during cancer progression. Several tumor microenvironment (TME)-derived factors orchestrate neutrophil release from the bone marrow, recruitment and functional polarization, while simultaneously neutrophils are active stimulators of the TME by secreting factors that affect immune interactions and subsequently tumor progression. Successful immunotherapies for many cancer types and stages depend on the targeting of tumor-infiltrating lymphocytes. Neutrophils impact the success of immunotherapies, such as immune checkpoint blockade therapies, by displaying lymphocyte suppressive properties. The identification and characterization of distinct neutrophil subpopulations or polarization states with pro- and antitumor phenotypes and the identification of the major TME-derived factors of neutrophil polarization would allow us to harness the full potential of neutrophils as complementary targets in anticancer precision therapies.
Collapse
Affiliation(s)
| | | | | | | | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (S.R.); (P.V.-C.); (Z.N.M.); (R.S.)
| |
Collapse
|
12
|
Singh P, Yadav M, Niveria K, Verma AK. Nano-immunotherapeutics: targeting approach as strategic regulation at tumor microenvironment for cancer treatment. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancer is the leading cause of mortality worldwide, which necessitates our consideration related to novel treatment approach. Tumor cells at the tumor microenvironment (TME), regulate a plethora of key mechanistic signaling pathways that obstruct antitumor immune responses by immune suppression, immune resistance or acquired immune tolerance. The present therapeutic regimes are provided independently or in combination, or as immunotherapies for cancer immune targeting. Immunotherapy has altered the arena of oncology and patient care. By using the host immune system, the immunostimulatory molecules can exert a robust, personalized response against the patient’s own tumors. Alternatively, tumors may exploit these strategies to escape immune recognition, and accordingly, such mechanisms represent chances for immunotherapy intervention. Nonetheless, despite promising outcomes from immunotherapies in recurrent and metastatic cancers, immune-therapeutics in clinics has been limited owing to unpredictability in the produced immune response and reported instances of immune-related adverse effects. The unrealized potential of immunotherapies in cancer management maybe due to the obstacles such as heterogeneous nature, multiple targets, patients’ immune response, specificity for cancer or variability in response generation in toxicity levels, delivery and cost related to therapeutics etc. Further revolutionary trends related to immunotherapies are noticeable with slower progress for cancer management. Recent advances in nanomedicine strategize to ameliorate the lacuna of immunotherapy as it relies on the inherent biophysical characteristics of nanocarriers: size, shape, surface charge and multifunctionality and exploiting them as first line therapy for delivery of biomolecules, single checkpoint inhibitors and for imaging of TME. Therefore, nano-assisted immunotherapies can boost the immunotherapeutic approach, overcoming factors that are with imminent potential risks related to it, thereby significantly improving the survival rate associated with it in cancer patients. Nanotechnology is anticipated to overcome the confines of existing cancer immunotherapy and to successfully combine various cancer treatment modes.
Collapse
Affiliation(s)
- Priyanka Singh
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi 110007, India
| | - Monika Yadav
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi 110007, India
| | - Karishma Niveria
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi 110007, India
| | - Anita Kamra Verma
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi 110007, India
| |
Collapse
|
13
|
BRAF and MEK Inhibitors Affect Dendritic-Cell Maturation and T-Cell Stimulation. Int J Mol Sci 2021; 22:ijms222111951. [PMID: 34769379 PMCID: PMC8585071 DOI: 10.3390/ijms222111951] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022] Open
Abstract
BRAF and MEK inhibitor (BRAFi/MEKi) combinations are currently the standard treatment for patients with BRAFV600 mutant metastatic melanoma. Since the RAS/RAF/MEK/ERK-pathway is crucial for the function of different immune cells, we postulated an effect on their function and thus interference with anti-tumor immunity. Therefore, we examined the influence of BRAFi/MEKi, either as single agent or in combination, on the maturation of monocyte-derived dendritic cells (moDCs) and their interaction with T cells. DCs matured in the presence of vemurafenib or vemurafenib/cobimetinib altered their cytokine secretion and surface marker expression profile. Upon the antigen-specific stimulation of CD8+ and CD4+ T cells with these DCs or with T2.A1 cells in the presence of BRAFi/MEKi, we detected a lower expression of activation markers on and a lower cytokine secretion by these T cells. However, treatment with any of the inhibitors alone or in combination did not change the avidity of CD8+ T cells in peptide titration assays with T2.A1 cells. T-helper cell/DC interaction is a bi-directional process that normally results in DC activation. Vemurafenib and vemurafenib/cobimetinib completely abolished the helper T-cell-mediated upregulation of CD70, CD80, and CD86 but not CD25 on the DCs. The combination of dabrafenib/trametinib affected DC maturation and activation as well as T-cell activation less than combined vemurafenib/cobimetinib did. Hence, for a potential combination with immunotherapy, our data indicate the superiority of dabrafenib/trametinib treatment.
Collapse
|
14
|
Stege H, Haist M, Nikfarjam U, Schultheis M, Heinz J, Pemler S, Loquai C, Grabbe S. The Status of Adjuvant and Neoadjuvant Melanoma Therapy, New Developments and Upcoming Challenges. Target Oncol 2021; 16:537-552. [PMID: 34554353 PMCID: PMC8484171 DOI: 10.1007/s11523-021-00840-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 12/14/2022]
Abstract
The global incidence of malignant melanoma, the leading cause of skin cancer death, has steadily increased in recent years. Surgical excision is the treatment of choice for early-stage melanoma. However, 40-60% of patients with high-risk melanoma or with nodal involvement eventually experience loco-regional relapse or tumor progression. Adjuvant therapy aims to reduce the rate of recurrence in radically operated high-risk patients with melanoma and thus improves survival. Interferon-α has long been the only approved drug for adjuvant melanoma therapy, despite an unclear survival benefit. The landmark success of immune-checkpoint inhibitors and BRAF/MEK-directed targeted therapies in the treatment of patients with stage IV melanoma led to the initiation of clinical trials in the adjuvant setting. These trials demonstrated the efficacy of immune-checkpoint inhibitors and targeted therapies for the adjuvant treatment of high-risk patients with melanoma, as shown both by an increase in recurrence-free survival and the emergence of long-term survivors, finally resulting in the approval of the cytotoxic T-lymphocyte antigen 4 inhibitor ipilimumab, PD1 inhibitors (nivolumab, pembrolizumab), and BRAF/MEK inhibitors for adjuvant melanoma therapy. This review aims to delineate the advances in adjuvant melanoma therapy, issuing particularly recent results from clinical trials. Moreover, we also discuss pending issues and future challenges, which comprise the adequate selection of adjuvant regimens for patient subgroups and the identification of markers likely to predict the individual response to adjuvant treatments. Last, we outline the role of emerging neoadjuvant approaches, which may complement adjuvant strategies and are currently investigated in clinical trials.
Collapse
Affiliation(s)
- Henner Stege
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Maximilian Haist
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ulrike Nikfarjam
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael Schultheis
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jaqueline Heinz
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Saskia Pemler
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
15
|
Katsandris A, Ziogas DC, Kontouri M, Staikoglou S, Gogas H. Atezolizumab plus vemurafenib and cobimetinib for the treatment of BRAF V600-mutant advanced melanoma: from an hypothetic triplet to an approved regimen. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1976637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Aikaterini Katsandris
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Dimitrios C. Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Maria Kontouri
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Stavroula Staikoglou
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| |
Collapse
|
16
|
Jung T, Haist M, Kuske M, Grabbe S, Bros M. Immunomodulatory Properties of BRAF and MEK Inhibitors Used for Melanoma Therapy-Paradoxical ERK Activation and Beyond. Int J Mol Sci 2021; 22:ijms22189890. [PMID: 34576054 PMCID: PMC8469254 DOI: 10.3390/ijms22189890] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/21/2022] Open
Abstract
The advent of mitogen-activated protein kinase (MAPK) inhibitors that directly inhibit tumor growth and of immune checkpoint inhibitors (ICI) that boost effector T cell responses have strongly improved the treatment of metastatic melanoma. In about half of all melanoma patients, tumor growth is driven by gain-of-function mutations of BRAF (v-rat fibrosarcoma (Raf) murine sarcoma viral oncogene homolog B), which results in constitutive ERK activation. Patients with a BRAF mutation are regularly treated with a combination of BRAF and MEK (MAPK/ERK kinase) inhibitors. Next to the antiproliferative effects of BRAF/MEKi, accumulating preclinical evidence suggests that BRAF/MEKi exert immunomodulatory functions such as paradoxical ERK activation as well as additional effects in non-tumor cells. In this review, we present the current knowledge on the immunomodulatory functions of BRAF/MEKi as well as the non-intended effects of ICI and discuss the potential synergistic effects of ICI and MAPK inhibitors in melanoma treatment.
Collapse
|
17
|
Babačić H, Eriksson H, Pernemalm M. Plasma proteome alterations by MAPK inhibitors in BRAF V600-mutated metastatic cutaneous melanoma. Neoplasia 2021; 23:783-791. [PMID: 34246984 PMCID: PMC8274243 DOI: 10.1016/j.neo.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/22/2022]
Abstract
Approximately half of metastatic cutaneous melanomas (CM) harbor a mutation in the BRAF protooncogene, upregulating the mitogen-activated protein kinase (MAPK)-pathway. The development of inhibitors targeting the MAPK pathway (MAPKi), i.e., BRAF- and MEK-inhibitors (BRAFi and MEKi), have substantially improved the survival in BRAFV600E/K-mutated stage IV metastatic CM. However, most patients develop resistance to treatment and no predictive biomarkers exist in practice. This study aimed at discovering plasma proteome changes during treatment MAPKi in patients with metastatic (stage IV) CM. Matched plasma samples before (pre) and during treatment (trm) from 23 patients with stage IV CM, treated with BRAF-inhibitors (BRAFi) alone or BRAF- and MEK- inhibitors combined (BRAFi and MEKi), were collected and analyzed with targeted proteomics by proximity extension assays. Additionally, plasma from 9 patients treated with BRAFi and MEKi was analyzed with in-depth high-resolution isoelectric focusing liquid-chromatography mass-spectrometry proteomics. Alterations of plasma proteins involved in granzyme and interferon gamma pathways were detected in patients treated with BRAFi, and cell adhesion-, neutrophil degranulation-, and proteolysis pathways in patients treated with BRAFi and MEKi. Several proteins were associated with progression-free survival after MAPKi treatment. We show that the majority of the altered plasma proteins were traceable to BRAFV600E-mutant metastatic CM tissue at mRNA level in 154 patients from the TCGA, further strengthening their involvement in tumoral response to treatment. This wide screen of plasma proteins unravels proteins that may serve as predictive and/or prognostic biomarkers of MAPKi treatment, opening a window of opportunity for plasma biomarker discovery in MAPKi-treatment of BRAFV600-mutant metastatic CM.
Collapse
Affiliation(s)
- Haris Babačić
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Hanna Eriksson
- Theme Cancer / Department of Oncology, Karolinska University Hospital, Stockholm, Sweden.
| | - Maria Pernemalm
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
18
|
Ziogas DC, Konstantinou F, Bouros S, Theochari M, Gogas H. Combining BRAF/MEK Inhibitors with Immunotherapy in the Treatment of Metastatic Melanoma. Am J Clin Dermatol 2021; 22:301-314. [PMID: 33765322 DOI: 10.1007/s40257-021-00593-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
The management and prognosis of BRAF-mutant metastatic melanoma have changed drastically following the introduction of immune checkpoint inhibitors and molecularly targeted agents. These treatment options present different mechanisms of action and toxicities but also totally distinct kinetics of their response, including a "relatively" short-lasting benefit in subsets of patients treated with BRAF/MEK inhibitors and a lower response rate in patients treated with immune checkpoint inhibitors. BRAF/MEK inhibitors, when administered prior to or concurrently with immune checkpoint inhibitors, at least transiently alter some immunosuppressive parameters of the tumor microenvironment and theoretically improve sensitivity to immunotherapy. Preclinical data from mouse models with oncogene-addicted melanoma confirmed this beneficial immune/targeted synergy and supported the clinical testing of combinations of BRAF/MEK inhibitors and immune checkpoint inhibitors to improve the activity of upfront anti-melanoma therapies. The first positive phase III results were published in 2020, and triggered the discussion about the benefits, the limitations, as well as the possible implications of combining or sequencing targeted therapies with immune checkpoint inhibitors in everyday practice. Beginning from the interplay of immune/targeted agents within the melanoma microenvironment, this review outlines available information from the retrospective experience up to the late-stage randomized evidence on combinatorial treatments. Many clinical trials are currently underway exploring open questions about optimal timing, new immune biomarkers, and eligible patient subsets for these immune/targeted regimens. Awaiting these results, decision making in the first-line setting for BRAF-mutant melanoma is still guided by the patients' characteristics and the biological aspects of melanoma.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Frosso Konstantinou
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Spyros Bouros
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Maria Theochari
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece
| | - Helen Gogas
- School of Medicine, First Department of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 75, Mikras Asias str., Goudi, 11527, Athens, Greece.
| |
Collapse
|
19
|
Peiffer L, Farahpour F, Sriram A, Spassova I, Hoffmann D, Kubat L, Stoitzner P, Gambichler T, Sucker A, Ugurel S, Schadendorf D, Becker JC. BRAF and MEK inhibition in melanoma patients enables reprogramming of tumor infiltrating lymphocytes. Cancer Immunol Immunother 2021; 70:1635-1647. [PMID: 33275172 PMCID: PMC8139931 DOI: 10.1007/s00262-020-02804-4] [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: 05/05/2020] [Accepted: 11/16/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Combined inhibition of BRAF/MEK is an established therapy for melanoma. In addition to its canonical mode of action, effects of BRAF/MEK inhibitors on antitumor immune responses are emerging. Thus, we investigated the effect of these on adaptive immune responses. PATIENTS, METHODS AND RESULTS Sequential tumor biopsies obtained before and during BRAF/MEK inhibitor treatment of four (n = 4) melanoma patients were analyzed. Multiplexed immunofluorescence staining of tumor tissue revealed an increased infiltration of CD4+ and CD8+ T cells upon therapy. Determination of the T-cell receptor repertoire usage demonstrated a therapy induced increase in T-cell clonotype richness and diversity. Application of the Grouping of Lymphocyte Interactions by Paratope Hotspots algorithm revealed a pre-existing immune response against melanoma differentiation and cancer testis antigens that expanded preferentially upon therapy. Indeed, most of the T-cell clonotypes found under BRAF/MEK inhibition were already present in lower numbers before therapy. This expansion appears to be facilitated by induction of T-bet and TCF7 in T cells, two transcription factors required for self-renewal and persistence of CD8+ memory T cells. CONCLUSIONS Our results suggest that BRAF/MEK inhibition in melanoma patients allows an increased expansion of pre-existing melanoma-specific T cells by induction of T-bet and TCF7 in these.
Collapse
Affiliation(s)
- Lukas Peiffer
- grid.5718.b0000 0001 2187 5445Deutsches Konsortium Für Translationale Krebsforschung (DKTK), Partner Site Essen, Translational Skin Cancer Research, University of Duisburg-Essen, Universitätsstr. 1, 45141 Essen, Germany ,grid.7497.d0000 0004 0492 0584Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Farnoush Farahpour
- grid.5718.b0000 0001 2187 5445Bioinformatics and Computational Biophysics, University Duisburg-Essen, Essen, Germany
| | - Ashwin Sriram
- grid.5718.b0000 0001 2187 5445Deutsches Konsortium Für Translationale Krebsforschung (DKTK), Partner Site Essen, Translational Skin Cancer Research, University of Duisburg-Essen, Universitätsstr. 1, 45141 Essen, Germany ,grid.7497.d0000 0004 0492 0584Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Ivelina Spassova
- grid.5718.b0000 0001 2187 5445Deutsches Konsortium Für Translationale Krebsforschung (DKTK), Partner Site Essen, Translational Skin Cancer Research, University of Duisburg-Essen, Universitätsstr. 1, 45141 Essen, Germany
| | - Daniel Hoffmann
- grid.5718.b0000 0001 2187 5445Bioinformatics and Computational Biophysics, University Duisburg-Essen, Essen, Germany
| | - Linda Kubat
- grid.5718.b0000 0001 2187 5445Deutsches Konsortium Für Translationale Krebsforschung (DKTK), Partner Site Essen, Translational Skin Cancer Research, University of Duisburg-Essen, Universitätsstr. 1, 45141 Essen, Germany
| | - Patrizia Stoitzner
- grid.5361.10000 0000 8853 2677Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thilo Gambichler
- grid.5570.70000 0004 0490 981XDepartment of Dermatology, Skin Cancer Center, Ruhr-University Bochum, Bochum, Germany
| | - Antje Sucker
- grid.410718.b0000 0001 0262 7331Department of Dermatology, University Hospital of Essen, Essen, Germany
| | - Selma Ugurel
- grid.410718.b0000 0001 0262 7331Department of Dermatology, University Hospital of Essen, Essen, Germany
| | - Dirk Schadendorf
- grid.410718.b0000 0001 0262 7331Department of Dermatology, University Hospital of Essen, Essen, Germany
| | - Jürgen C. Becker
- grid.5718.b0000 0001 2187 5445Deutsches Konsortium Für Translationale Krebsforschung (DKTK), Partner Site Essen, Translational Skin Cancer Research, University of Duisburg-Essen, Universitätsstr. 1, 45141 Essen, Germany ,grid.7497.d0000 0004 0492 0584Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany ,grid.410718.b0000 0001 0262 7331Department of Dermatology, University Hospital of Essen, Essen, Germany
| |
Collapse
|
20
|
Dummer R, Ascierto PA, Nathan P, Robert C, Schadendorf D. Rationale for Immune Checkpoint Inhibitors Plus Targeted Therapy in Metastatic Melanoma: A Review. JAMA Oncol 2020; 6:1957-1966. [PMID: 32970096 DOI: 10.1001/jamaoncol.2020.4401] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance In recent years, the management of metastatic melanoma has been transformed by the emergence of immune checkpoint inhibitors and targeted therapies that significantly improve patient survival. The complementary response kinetics of these treatment approaches, supported by mechanistic evidence that targeted therapy affects immune aspects of the tumor microenvironment, suggest that the optimal combination or sequencing of immune checkpoint inhibitors and targeted therapy may provide additional clinical benefit. Observations Clinical responses to BRAF and/or MEK inhibitors are associated with immune changes within the tumor microenvironment that have the potential to increase the sensitivity of BRAF V600-mutant melanoma to immune checkpoint inhibitors. The combination of immune checkpoint inhibitors with targeted therapy may therefore increase duration of response, improve tumor control, extend survival, and increase the proportion of patients experiencing durable benefit. A targeted therapy-immune checkpoint inhibitor sequencing approach may also be supported by this evidence, but clinical questions regarding optimal timing, duration, and patient selection remain. Conclusions and Relevance This review outlines the rationale and preclinical evidence that support immune checkpoint inhibitor plus targeted therapy combination and sequencing strategies in melanoma and highlights the results available to date from clinical trials exploring these approaches to treatment. Several late-stage trials are under way looking to answer open questions in this field and address the continuing debate surrounding up-front combination vs sequencing. As phase 3 data have begun to emerge, trial designs and available data from key studies are discussed in the context of their resultant implications for clinical practice.
Collapse
Affiliation(s)
- Reinhard Dummer
- University Hospital Zürich Skin Cancer Center, Zürich, Switzerland
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale," Naples, Italy
| | - Paul Nathan
- Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - Caroline Robert
- Gustave Roussy and Paris-Sud-Paris-Saclay University, Villejuif, France
| | - Dirk Schadendorf
- University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
21
|
Simiczyjew A, Dratkiewicz E, Mazurkiewicz J, Ziętek M, Matkowski R, Nowak D. The Influence of Tumor Microenvironment on Immune Escape of Melanoma. Int J Mol Sci 2020; 21:E8359. [PMID: 33171792 PMCID: PMC7664679 DOI: 10.3390/ijms21218359] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
The low efficiency of currently-used anti-cancer therapies poses a serious challenge, especially in the case of malignant melanoma, a cancer characterized by elevated invasiveness and relatively high mortality rate. The role of the tumor microenvironment in the progression of melanoma and its acquisition of resistance to treatment seems to be the main focus of recent studies. One of the factors that, in normal conditions, aids the organism in its fight against the cancer and, following the malignant transformation, adapts to facilitate the development of the tumor is the immune system. A variety of cell types, i.e., T and B lymphocytes, macrophages, and dendritic and natural killer cells, as well as neutrophils, support the growth and invasiveness of melanoma cells, utilizing a plethora of mechanisms, including secretion of pro-inflammatory molecules, induction of inhibitory receptors expression, or depletion of essential nutrients. This review provides a comprehensive summary of the processes regulated by tumor-associated cells that promote the immune escape of melanoma cells. The described mechanisms offer potential new targets for anti-cancer treatment and should be further studied to improve currently-employed therapies.
Collapse
Affiliation(s)
- Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (E.D.); (J.M.); (D.N.)
| | - Ewelina Dratkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (E.D.); (J.M.); (D.N.)
| | - Justyna Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (E.D.); (J.M.); (D.N.)
| | - Marcin Ziętek
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wroclaw, Poland; (M.Z.); (R.M.)
- Wroclaw Comprehensive Cancer Center, Plac Hirszfelda 12, 53-413 Wroclaw, Poland
| | - Rafał Matkowski
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wroclaw, Poland; (M.Z.); (R.M.)
- Wroclaw Comprehensive Cancer Center, Plac Hirszfelda 12, 53-413 Wroclaw, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (E.D.); (J.M.); (D.N.)
| |
Collapse
|
22
|
Zagorulya M, Duong E, Spranger S. Impact of anatomic site on antigen-presenting cells in cancer. J Immunother Cancer 2020; 8:e001204. [PMID: 33020244 PMCID: PMC7537336 DOI: 10.1136/jitc-2020-001204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 12/24/2022] Open
Abstract
Checkpoint blockade immunotherapy (CBT) can induce long-term clinical benefits in patients with advanced cancer; however, response rates to CBT vary by cancer type. Cancers of the skin, lung, and kidney are largely responsive to CBT, while cancers of the pancreas, ovary, breast, and metastatic lesions to the liver respond poorly. The impact of tissue-resident immune cells on antitumor immunity is an emerging area of investigation. Recent evidence indicates that antitumor immune responses and efficacy of CBT depend on the tissue site of the tumor lesion. As myeloid cells are predominantly tissue-resident and can shape tumor-reactive T cell responses, it is conceivable that tissue-specific differences in their function underlie the tissue-site-dependent variability in CBT responses. Understanding the roles of tissue-specific myeloid cells in antitumor immunity can open new avenues for treatment design. In this review, we discuss the roles of tissue-specific antigen-presenting cells (APCs) in governing antitumor immune responses, with a particular focus on the contributions of tissue-specific dendritic cells. Using the framework of the Cancer-Immunity Cycle, we examine the contributions of tissue-specific APC in CBT-sensitive and CBT-resistant carcinomas, highlight how these cells can be therapeutically modulated, and identify gaps in knowledge that remain to be addressed.
Collapse
Affiliation(s)
- Maria Zagorulya
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ellen Duong
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Stefani Spranger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
23
|
Abstract
Kinases form the major part of the druggable genome and their selective inhibition in human cancers has had reasonable clinical success. In contrast to tumorigenesis, the role of kinases in mediating immune responses is poorly understood. However, synergistic therapeutic regimens combining targeted therapy and immune therapy have been found to increase the median survival of tumor patients. In this context, we uncovered that RAF and MEK1/2 kinases, which are the integral parts of the classical MAPK cascade, have unique roles in driving DC differentiation and activation. RAF kinases are stabilized in their protein levels during DC differentiation and are obligatory for normal functioning of DCs. But, the targeting of MEK1/2 kinases with specific inhibitors did not phenocopy the effects observed with RAF inhibitors suggesting that RAF and MEK1/2 kinases may have specific and unique roles in driving immune responses, which deserves further studies to successfully administer these inhibitors in clinics.
Collapse
Affiliation(s)
- Kristina Riegel
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz , Mainz, Germany
| | | |
Collapse
|
24
|
Monti M, Vescovi R, Consoli F, Farina D, Moratto D, Berruti A, Specchia C, Vermi W. Plasmacytoid Dendritic Cell Impairment in Metastatic Melanoma by Lactic Acidosis. Cancers (Basel) 2020; 12:cancers12082085. [PMID: 32731406 PMCID: PMC7463681 DOI: 10.3390/cancers12082085] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
The introduction of targeted therapies and immunotherapies has significantly improved the outcome of metastatic melanoma (MM) patients. These approaches rely on immune functions for their anti-melanoma response. Plasmacytoid dendritic cells (pDCs) exhibit anti-tumor function by production of effector molecules, type I interferons (I-IFNs), and cytokines. Tissue and blood pDCs result compromised in MM, although these findings are still partially conflicting. This study reports that blood pDCs were dramatically depleted in MM, particularly in patients with high lactate dehydrogenase (LDH) and high tumor burden; the reduced pDC frequency was associated with poor overall survival. Circulating pDCs resulted also in significant impairment in interferon alpha (IFN-α) and C-X-C motif chemokine 10 (CXCL10) production in response to toll-like receptor (TLR)-7/8 agonists; on the contrary, the response to TLR-9 agonist remained intact. In the BRAFV600+ subgroup, no recovery of pDC frequency could be obtained by BRAF and MEK inhibitors (BRAFi; MEKi), whereas their function was partially rescued. Mechanistically, in vitro exposure to lactic acidosis impaired both pDC viability and function. In conclusion, pDCs from MM patients were found to be severely impaired, with a potential role for lactic acidosis. Short-term responses to treatments were not associated with pDC recovery, suggesting long-lasting effects on their compartment.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (C.S.)
| | - Raffaella Vescovi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (C.S.)
| | - Francesca Consoli
- Oncology Unit, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (F.C.); (A.B.)
| | - Davide Farina
- Radiology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25123 Brescia, Italy;
| | - Daniele Moratto
- Laboratory of Genetic Disorders of Childhood, Angelo Nocivelli Institute for Molecular Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
| | - Alfredo Berruti
- Oncology Unit, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (F.C.); (A.B.)
| | - Claudia Specchia
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (C.S.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (C.S.)
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63101, USA
- Correspondence: ; Tel.: +39-030-399-8425
| |
Collapse
|
25
|
Proietti I, Skroza N, Michelini S, Mambrin A, Balduzzi V, Bernardini N, Marchesiello A, Tolino E, Volpe S, Maddalena P, Di Fraia M, Mangino G, Romeo G, Potenza C. BRAF Inhibitors: Molecular Targeting and Immunomodulatory Actions. Cancers (Basel) 2020; 12:cancers12071823. [PMID: 32645969 PMCID: PMC7408709 DOI: 10.3390/cancers12071823] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
The BRAF inhibitors vemurafenib, dabrafenib and encorafenib are used in the treatment of patients with BRAF-mutant melanoma. They selectively target BRAF kinase and thus interfere with the mitogen-activated protein kinase (MAPK) signalling pathway that regulates the proliferation and survival of melanoma cells. In addition to their molecularly targeted activity, BRAF inhibitors have immunomodulatory effects. The MAPK pathway is involved in T-cell receptor signalling, and interference in the pathway by BRAF inhibitors has beneficial effects on the tumour microenvironment and anti-tumour immune response in BRAF-mutant melanoma, including increased immune-stimulatory cytokine levels, decreased immunosuppressive cytokine levels, enhanced melanoma differentiation antigen expression and presentation of tumour antigens by HLA 1, and increased intra-tumoral T-cell infiltration and activity. These effects promote recognition of the tumour by the immune system and enhance anti-tumour T-cell responses. Combining BRAF inhibitors with MEK inhibitors provides more complete blockade of the MAPK pathway. The immunomodulatory effects of BRAF inhibition alone or in combination with MEK inhibition provide a rationale for combining these targeted therapies with immune checkpoint inhibitors. Available data support the synergy between these treatment approaches, indicating such combinations provide an additional beneficial effect on the tumour microenvironment and immune response in BRAF-mutant melanoma.
Collapse
Affiliation(s)
- Ilaria Proietti
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
- Correspondence: ; Tel.: +39-3334684342 or +39-0773708811
| | - Nevena Skroza
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Simone Michelini
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Alessandra Mambrin
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Veronica Balduzzi
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Nicoletta Bernardini
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Anna Marchesiello
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Ersilia Tolino
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Salvatore Volpe
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Patrizia Maddalena
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Marco Di Fraia
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Giorgio Mangino
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.M.); (G.R.)
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00100 Rome, Italy
| | - Giovanna Romeo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.M.); (G.R.)
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00100 Rome, Italy
| | - Concetta Potenza
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| |
Collapse
|
26
|
Wang P, Zhang X, Sun N, Zhao Z, He J. Comprehensive Analysis of the Tumor Microenvironment in Cutaneous Melanoma associated with Immune Infiltration. J Cancer 2020; 11:3858-3870. [PMID: 32328190 PMCID: PMC7171484 DOI: 10.7150/jca.44413] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/27/2020] [Indexed: 12/25/2022] Open
Abstract
Accumulating evidence suggests that the malignant phenotypes of cancers are determined not only by the intrinsic properties of cancer cells but also by components in the tumor microenvironment (TME). In this study, we comprehensively characterized the TME of cutaneous melanoma (CM). As a result, tumor stage, tissue site, ulceration, thickness as well as patient age, sex were associated with immune infiltration. Patients of higher immune infiltration exhibited better survival outcomes, and antitumor effector cells, such as CD8 T cells and M1 macrophages, were found in significantly higher numbers in those tissues. Differential expression of mRNAs and long non-coding RNAs (lncRNAs) was analyzed and utilized to construct an immune-related competing endogenous RNA network, in which a lncRNA-associated subnetwork that could positively regulate the expression of IFN-γ was highlighted. Functional analysis confirmed that this network was remarkably enriched in functional terms related to both immune response and tumor-intrinsic pathways. Finally, a total of 109 high-confidence prognostic genes were identified, and a gene module that contained several key immune checkpoint molecules or modulators (PD-1, PD-L1, PD-L2, and LCK) was screened, which confers survival benefit for CM patients as supported by both overall and relapse-free survival rates from different datasets.
Collapse
Affiliation(s)
- Pan Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xinyu Zhang
- Department of body contouring and liposuction center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhihong Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
27
|
Monti M, Consoli F, Vescovi R, Bugatti M, Vermi W. Human Plasmacytoid Dendritic Cells and Cutaneous Melanoma. Cells 2020; 9:E417. [PMID: 32054102 PMCID: PMC7072514 DOI: 10.3390/cells9020417] [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: 12/24/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
The prognosis of metastatic melanoma (MM) patients has remained poor for a long time. However, the recent introduction of effective target therapies (BRAF and MEK inhibitors for BRAFV600-mutated MM) and immunotherapies (anti-CTLA-4 and anti-PD-1) has significantly improved the survival of MM patients. Notably, all these responses are highly dependent on the fitness of the host immune system, including the innate compartment. Among immune cells involved in cancer immunity, properly activated plasmacytoid dendritic cells (pDCs) exert an important role, bridging the innate and adaptive immune responses and directly eliminating cancer cells. A distinctive feature of pDCs is the production of high amount of type I Interferon (I-IFN), through the Toll-like receptor (TLR) 7 and 9 signaling pathway activation. However, published data indicate that melanoma-associated escape mechanisms are in place to hijack pDC functions. We have recently reported that pDC recruitment is recurrent in the early phases of melanoma, but the entire pDC compartment collapses over melanoma progression. Here, we summarize recent advances on pDC biology and function within the context of melanoma immunity.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Francesca Consoli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology, University of Brescia at ASST-Spedali Civili, 25123 Brescia, Italy;
| | - Raffaella Vescovi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
28
|
Friedman A, Siewe N. Overcoming Drug Resistance to BRAF Inhibitor. Bull Math Biol 2020; 82:8. [PMID: 31933021 DOI: 10.1007/s11538-019-00691-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/20/2019] [Indexed: 11/25/2022]
Abstract
One of the most frequently found mutations in human melanomas is in the B-raf gene, making its protein BRAF a key target for therapy. However, in patients treated with BRAF inhibitor (BRAFi), although the response is very good at first, relapse occurs within 6 months, on the average. In order to overcome this drug resistance to BRAFi, various combinations of BRAFi with other drugs have been explored, and some are being applied clinically, such as a combination of BRAF and MEK inhibitors. Experimental data for melanoma in mice show that under continuous treatment with BRAFi, the pro-cancer MDSCs and chemokine CCL2 initially decrease but eventually increase to above their original level, while the anticancer T cells continuously decrease. In this paper, we develop a mathematical model that explains these experimental results. The model is used to explore the efficacy of combinations of BRAFi with anti-CCL2, anti-PD-1 and anti-CTLA-4, with the aim of eliminating or reducing drug resistance to BRAFi.
Collapse
Affiliation(s)
- Avner Friedman
- Mathematical Biosciences Institute & Department of Mathematics, The Ohio State University, Columbus, OH, USA
| | - Nourridine Siewe
- Department of Mathematics, The University of British Columbia Okanagan, Kelowna, BC, Canada.
| |
Collapse
|
29
|
Ziogas DC, Konstantinou F, Bouros S, Gogas H. Identifying the optimum first-line therapy in BRAF-mutant metastatic melanoma. Expert Rev Anticancer Ther 2020; 20:53-62. [PMID: 31903803 DOI: 10.1080/14737140.2020.1711737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: The emergence of molecularly targeted agents and immune checkpoint inhibitors has positively revolutionized the management and prognosis of BRAF-mutant metastatic melanoma. However, the availability of both therapeutic options, with their pros and cons, rationally triggered clinical considerations for the optimum frontline and subsequent treatment decisions.Areas covered: Here, we debate all approved therapies in patients with BRAF-mutant metastatic melanoma evaluating their efficacy and safety based on their pivotal trials. With prospective randomized data pending, retrospective comparisons of BRAF/MEK versus immune checkpoint inhibitors are reviewed to recognize any advantage between these two alternatives and to optimize their implementation. Preclinical and early clinical results of combining concurrently or sequentially targeted therapy and immunotherapy are also discussed.Expert opinion: BRAF/MEK inhibitors produce rapid and deep responses and should be included in first-line approaches, particularly in cases with aggressive and bulky disease, while single or double checkpoint inhibition lead to more durable responses and could be involved either in frontline treatment of BRAF-mutant melanoma with less unfavorable characteristics or in maintenance after initial targeted induction or in future immune/targeted regimens for high-risk groups. Data from ongoing trials directly comparing or combining these strategies are expected to update their role in a more individualized basis.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Frosso Konstantinou
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Spyros Bouros
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| |
Collapse
|
30
|
Luís R, Brito C, Pojo M. Melanoma Metabolism: Cell Survival and Resistance to Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:203-223. [PMID: 32130701 DOI: 10.1007/978-3-030-34025-4_11] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cutaneous melanoma is one of the most aggressive types of cancer, presenting the highest potential to form metastases, both locally and distally, which are associated with high death rates of melanoma patients. A high somatic mutation burden is characteristic of these tumours, with most common oncogenic mutations occurring in the BRAF, NRAS and NF1 genes. These intrinsic oncogenic pathways contribute to the metabolic switch between glycolysis and oxidative phosphorylation metabolisms of melanoma, facilitating tumour progression and resulting in a high plasticity and adaptability to unfavourable conditions. Moreover, melanoma microenvironment can influence its own metabolism and reprogram several immune cell subset functions, enabling melanoma to evade the immune system. The knowledge of the biology, molecular alterations and microenvironment of melanoma has led to the development of new targeted therapies and the improvement of patient care. In this work, we reviewed the impact of melanoma metabolism in the resistance to BRAF and MEK inhibitors and immunotherapies, emphasizing the requirement to evaluate metabolic alterations upon development of novel therapeutic approaches. Here we summarized the current understanding of the impact of metabolic processes in melanomagenesis, metastasis and microenvironment, as well as the involvement of metabolic pathways in the immune modulation and resistance to targeted and immunocheckpoint therapies.
Collapse
Affiliation(s)
- Rafael Luís
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| | - Cheila Brito
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| | - Marta Pojo
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| |
Collapse
|
31
|
Yang L, Li A, Lei Q, Zhang Y. Tumor-intrinsic signaling pathways: key roles in the regulation of the immunosuppressive tumor microenvironment. J Hematol Oncol 2019; 12:125. [PMID: 31775797 PMCID: PMC6880373 DOI: 10.1186/s13045-019-0804-8] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/02/2019] [Indexed: 12/17/2022] Open
Abstract
Immunotherapy is a currently popular treatment strategy for cancer patients. Although recent developments in cancer immunotherapy have had significant clinical impact, only a subset of patients exhibits clinical response. Therefore, understanding the molecular mechanisms of immunotherapy resistance is necessary. The mechanisms of immune escape appear to consist of two distinct tumor characteristics: a decrease in effective immunocyte infiltration and function and the accumulation of immunosuppressive cells in the tumor microenvironment. Several host-derived factors may also contribute to immune escape. Moreover, inter-patient heterogeneity predominantly results from differences in somatic mutations between cancers, which has led to the hypothesis that differential activation of specific tumor-intrinsic pathways may explain the phenomenon of immune exclusion in a subset of cancers. Increasing evidence has also shown that tumor-intrinsic signaling plays a key role in regulating the immunosuppressive tumor microenvironment and tumor immune escape. Therefore, understanding the mechanisms underlying immune avoidance mediated by tumor-intrinsic signaling may help identify new therapeutic targets for expanding the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, 450052, People's Republic of China
| | - Aitian Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, 450052, People's Republic of China
| | - Qingyang Lei
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, 450052, People's Republic of China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China. .,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China. .,School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China. .,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, 450052, People's Republic of China.
| |
Collapse
|
32
|
Croce L, Coperchini F, Magri F, Chiovato L, Rotondi M. The multifaceted anti-cancer effects of BRAF-inhibitors. Oncotarget 2019; 10:6623-6640. [PMID: 31762942 PMCID: PMC6859927 DOI: 10.18632/oncotarget.27304] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/19/2019] [Indexed: 12/26/2022] Open
Abstract
The BRAF gene is commonly involved in normal processes of cell growth and differentiation. The BRAF (V600E) mutation is found in several human cancer, causing an increase of cell proliferation due to a modification of the ERK/MAPK-signal cascade. In particular, BRAFV600E mutation is found in those melanoma or thyroid cancer refractory to the common therapy and with a more aggressive phenotype. BRAF V600E was found to influence the composition of the so-called tumour microenvironment modulating both solid (immune-cell infiltration) and soluble (chemokines) mediators, which balance characterize the ultimate behaviour of the tumour, making it more or less aggressive. In particular, the presence of BRAFV600E mutation would be associated with a change of this balance to a more aggressive phenotype of the tumour and a worse prognosis. The investigation of the possible modulation of those components of tumour microenvironment is nowadays object of several studies as a new potential target therapy in those more complicated cases. At present several clinical trials both in melanoma and thyroid cancer are using BRAF-inhibitors with encouraging results, which are derived also from numerous in vitro pre-clinical studies aimed at evaluate the possible modulation of immune-cell density and of specific pro-tumorigenic chemokine secretion (CXCL8 and CCL2) by several BRAF-inhibitors in the context of melanoma and thyroid cancer. This review will encompass in vitro and in vivo studies which investigated the modulation of the tumour microenvironment by BRAF-inhibitors, highlighting also the most recent clinical trials with a specific focus on melanoma and thyroid cancer.
Collapse
Affiliation(s)
- Laura Croce
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
- PHD course in Experimental Medicine, University of Pavia, Pavia, Italy
| | - Francesca Coperchini
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
| | - Flavia Magri
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Luca Chiovato
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Mario Rotondi
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| |
Collapse
|
33
|
Tucci M, Passarelli A, Mannavola F, Felici C, Stucci LS, Cives M, Silvestris F. Immune System Evasion as Hallmark of Melanoma Progression: The Role of Dendritic Cells. Front Oncol 2019; 9:1148. [PMID: 31750245 PMCID: PMC6848379 DOI: 10.3389/fonc.2019.01148] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
Melanoma is an immunogenic tumor whose relationship with immune cells resident in the microenvironment significantly influences cancer cell proliferation, progression, and metastasis. During melanomagenesis, both immune and melanoma cells undergo the immunoediting process that includes interconnected phases as elimination, equilibrium, and escape or immune evasion. In this context, dendritic cells (DCs) are active players that indirectly counteract the proliferation of melanoma cells. Moreover, DC maturation, migration, and cross-priming as well as their functional interplay with cytotoxic T-cells through ligands of immune checkpoint receptors result impaired. A number of signals propagated by highly proliferating melanoma cells and accessory cells as T-cells, natural killer cells (NKs), tumor-associated macrophages (TAMs), T-regulatory cells (T-regs), myeloid-derived suppressor cells (MDSCs), and endothelial cells participate to create an immunosuppressive milieu that results engulfed of tolerogenic factors and interleukins (IL) as IL-6 and IL-10. To underline the role of the immune infiltrate in blocking the melanoma progression, it has been described that the composition, density, and distribution of cytotoxic T-cells in the surrounding stroma is predictive of responsiveness to immunotherapy. Here, we review the major mechanisms implicated in melanoma progression, focusing on the role of DCs.
Collapse
Affiliation(s)
- Marco Tucci
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Anna Passarelli
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Mannavola
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Claudia Felici
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Luigia Stefania Stucci
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Mauro Cives
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Silvestris
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
34
|
RAF kinases are stabilized and required for dendritic cell differentiation and function. Cell Death Differ 2019; 27:1300-1315. [PMID: 31541179 PMCID: PMC7206131 DOI: 10.1038/s41418-019-0416-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/23/2019] [Accepted: 08/30/2019] [Indexed: 12/05/2022] Open
Abstract
RAF kinases (ARAF, BRAF, and CRAF) are highly conserved enzymes that trigger the RAF-MEK1/2-ERK1/2 (MAPK) pathway upon activation of RAS. Despite enormous clinical interest, relatively little is known on the role of RAFs in mediating immune responses. Here, we investigated the role of RAF kinases and MEK1/2 in dendritic cells (DCs), the central regulators of T cell-mediated antitumor immune responses and the adaptive immune system. We demonstrate that RAF kinases are active and stabilized at their protein levels during DC differentiation. Inhibition of RAF kinases but not MEK1/2 impaired the activation of DCs in both mice and human. As expected, DCs treated with RAF inhibitors show defects in activating T cells. Further, RAF and MEK1/2 kinases are directly required for the activation and proliferation of CD4+ T cells. Our observations suggest that RAF and MEK1/2 have independent roles in regulating DC function that has important implications for administering RAF–MAPK inhibitors in the clinics.
Collapse
|
35
|
Rossi A, Roberto M, Panebianco M, Botticelli A, Mazzuca F, Marchetti P. Drug resistance of BRAF-mutant melanoma: Review of up-to-date mechanisms of action and promising targeted agents. Eur J Pharmacol 2019; 862:172621. [PMID: 31446019 DOI: 10.1016/j.ejphar.2019.172621] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 12/29/2022]
Abstract
Melanoma onset and progression are associated with a high variety of activating mutations in the MAPK-pathway, most frequently involving BRAF (35-45%) and NRAS (15-25%) genes, but also c-KIT and PTEN. Targeted therapies with BRAF and MEK inhibitors showed promising results over the past years, but it is known that most responses are temporary, and almost all of patients develop a tumor relapse within one year. Different drug-resistance mechanisms underlie the progression of disease and activation of both MAPK and PI3K/AKT/mTOR pathways. Therefore, in this article we reviewed the main studies about clinical effects of several target inhibitors, describing properly the most prominent mechanisms of both intrinsic and acquired resistance. Furthermore, suggestive strategies for overcoming drug resistance and the most recent alternative combination therapies to optimize the use of MAPK pathway inhibitors were also discussed.
Collapse
Affiliation(s)
- Alessandro Rossi
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Michela Roberto
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy; Department of Medical-Surgical Sciences and Translation Medicine, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy.
| | - Martina Panebianco
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy; Oncology Unit, IDI-IRCCS of Rome, Italy
| |
Collapse
|
36
|
Yu C, Liu X, Yang J, Zhang M, Jin H, Ma X, Shi H. Combination of Immunotherapy With Targeted Therapy: Theory and Practice in Metastatic Melanoma. Front Immunol 2019; 10:990. [PMID: 31134073 PMCID: PMC6513976 DOI: 10.3389/fimmu.2019.00990] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/16/2019] [Indexed: 02/05/2023] Open
Abstract
Metastatic melanoma is the most aggressive and obstinate skin cancer with poor prognosis. Variant novel applicable regimens have emerged during the past decades intensively, while the most profound approaches are oncogene-targeted therapy and T-lymphocyte mediated immunotherapy. Although targeted therapies generated remarkable and rapid clinical responses in the majority of patients, acquired resistance was developed promptly within months leading to tumor relapse. By contrast, immunotherapies elicited long-term tumor regression. However, the overall response rate was limited. In view of the above, either targeted therapy or immunotherapy cannot elicit durable clinical responses in large range of patients. Interestingly, the advantages and limitations of these regimens happened to be complementary. An increasing number of preclinical studies and clinical trials proved a synergistic antitumor effect with the combination of targeted therapy and immunotherapy, implying a promising prospect for the treatment of metastatic melanoma. In order to achieve a better therapeutic effectiveness and reduce toxicity in patients, great efforts need to be made to illuminate multifaceted interplay between targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Chune Yu
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowei Liu
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiqiao Yang
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Min Zhang
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongyu Jin
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hubing Shi
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Sclafani F. MEK and PD-L1 inhibition in colorectal cancer: a burning blaze turning into a flash in the pan. Lancet Oncol 2019; 20:752-753. [PMID: 31003912 DOI: 10.1016/s1470-2045(19)30076-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 11/19/2022]
Affiliation(s)
- Francesco Sclafani
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet, Brussels 1000, Belgium; Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
38
|
Indini A, Tondini CA, Mandalà M. Cobimetinib in malignant melanoma: how to MEK an impact on long-term survival. Future Oncol 2019; 15:967-977. [DOI: 10.2217/fon-2018-0659] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Approximately 50% of cutaneous melanomas harbor activating mutations of the BRAF-oncogene, making BRAF inhibitors (BRAFi) the standard treatment for this disease. However, disease responses are limited in duration mainly due to acquired resistance. Dual MAPK pathway inhibition with addition of a MEK inhibitor (MEKi) to a BRAFi improved the efficacy and tolerability compared with BRAFi alone. Cobimetinib (Cotellic®) is an orally bioavailable, potent and selective MEKi, which significantly improved response rates when combined with BRAFi vemurafenib (median overall survival: 22.3 months). The toxicity profile of cobimetinib is manageable and treatment discontinuation due to adverse events is uncommon. Present efforts are addressed to overcome resistance and improve long-term outcomes: based on the evidence of the immunomodulatory properties of BRAFi and MEKi, current clinical trials of combined targeted and immunotherapy are investigating the role of cobimetinib in the context of combination or as sequential treatments.
Collapse
Affiliation(s)
- Alice Indini
- Unit of Medical Oncology, Department of Oncology & Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Carlo Alberto Tondini
- Unit of Medical Oncology, Department of Oncology & Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Mario Mandalà
- Unit of Medical Oncology, Department of Oncology & Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
39
|
Pelster MS, Amaria RN. Combined targeted therapy and immunotherapy in melanoma: a review of the impact on the tumor microenvironment and outcomes of early clinical trials. Ther Adv Med Oncol 2019; 11:1758835919830826. [PMID: 30815041 PMCID: PMC6384439 DOI: 10.1177/1758835919830826] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/23/2019] [Indexed: 12/12/2022] Open
Abstract
The development of BRAF and MEK inhibitors (BRAFis and MEKis) and immune
checkpoint inhibitors have changed the management of advanced stage melanoma and
improved the outcomes of patients with this malignancy. However, both
therapeutic approaches have limitations, including a limited duration of benefit
in subsets of BRAF-mutant melanoma patients treated with targeted therapy and a
lower overall response rate without a clear predictive biomarker in patients
treated with checkpoint inhibitors. Preclinical and translational data have
shown that BRAFis and MEKis alter the tumor microenvironment to make it more
amenable to immunotherapy and have provided the scientific rationale for combing
BRAFis and MEKis with immunotherapy. In this review, the initial studies
demonstrating the impact of BRAFis and MEKis on the expression of melanoma
differentiation antigens, T-cell infiltration, and the balance of immune
stimulatory and immune suppressive cells and cytokines are addressed.
Preclinical work on the combination of targeted therapy with BRAFis and MEKis
with immunotherapy are reviewed, highlighting improved tumor responses in mouse
models of BRAF-mutated melanoma treated with combinatorial strategies. Lastly,
data from early clinical trials of combined targeted therapy and immunotherapy
are discussed, focusing on response rates and toxicities.
Collapse
Affiliation(s)
- Meredith S Pelster
- The University of Texas MD Anderson Cancer Center, Department of Hematology/Oncology, Houston, TX, USA
| | - Rodabe N Amaria
- The University of Texas MD Anderson Cancer Center, Department of Melanoma Medical Oncology, 1515 Holcombe Boulevard, Unit 0430, Houston, TX 77030, USA
| |
Collapse
|
40
|
Awad RM, De Vlaeminck Y, Maebe J, Goyvaerts C, Breckpot K. Turn Back the TIMe: Targeting Tumor Infiltrating Myeloid Cells to Revert Cancer Progression. Front Immunol 2018; 9:1977. [PMID: 30233579 PMCID: PMC6127274 DOI: 10.3389/fimmu.2018.01977] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor cells frequently produce soluble factors that favor myelopoiesis and recruitment of myeloid cells to the tumor microenvironment (TME). Consequently, the TME of many cancer types is characterized by high infiltration of monocytes, macrophages, dendritic cells and granulocytes. Experimental and clinical studies show that most myeloid cells are kept in an immature state in the TME. These studies further show that tumor-derived factors mold these myeloid cells into cells that support cancer initiation and progression, amongst others by enabling immune evasion, tumor cell survival, proliferation, migration and metastasis. The key role of myeloid cells in cancer is further evidenced by the fact that they negatively impact on virtually all types of cancer therapy. Therefore, tumor-associated myeloid cells have been designated as the culprits in cancer. We review myeloid cells in the TME with a focus on the mechanisms they exploit to support cancer cells. In addition, we provide an overview of approaches that are under investigation to deplete myeloid cells or redirect their function, as these hold promise to overcome resistance to current cancer therapies.
Collapse
|
41
|
Immunomodulatory effects of BRAF and MEK inhibitors: Implications for Melanoma therapy. Pharmacol Res 2018; 136:151-159. [PMID: 30145328 DOI: 10.1016/j.phrs.2018.08.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/21/2022]
Abstract
Targeted therapy with BRAF inhibitors (BRAFi) and MEK inhibitors (MEKi) provides rapid disease control with high response rates in patients with BRAF-mutant metastatic melanoma. However, the majority of patients develop resistance to therapy during the course of therapy. Immune checkpoint inhibitors show a slower onset of action with lower response rates, with responders showing sustained response. The combination of BRAFi/MEKi and immune checkpoint inhibitors combines the hope for a fast, reliable and lasting response to therapy. Preclinical data supports this hypothesis. With the help of the PubMed database, a comprehensive search and analysis of preclinical and clinical studies on the combination of BRAFi/MEKi with immune checkpoint inhibitors was performed and yielded the following results: 1) In vivo, BRAFi and MEKi have no negative effects on immune cells; BRAFi and MEKi generate 2) an immune stimulating tumor microenvironment, 3) an increased infiltration of immune cells into the tumors, 4) a better recognition of melanoma cells by immune effector cells, and 5) a better functionality of the immune effector cells. In addition, in vivo experiments 6) demonstrated a superiority of the combination treatment compared to the individual strategies in both BRAF-mutant and BRAF wild-type melanomas. In summary, available data show that both BRAFi and MEKi have beneficial effects on the antitumor immunity and the tumor microenvironment as a whole, which is mediated by different mechanisms. Currently, clinical studies are underway to investigate combinations of BRAFi and MEKi with immune checkpoint inhibitors. The results of these studies are eagerly awaited.
Collapse
|
42
|
Hajek E, Krebs F, Bent R, Haas K, Bast A, Steinmetz I, Tuettenberg A, Grabbe S, Bros M. BRAF inhibitors stimulate inflammasome activation and interleukin 1 beta production in dendritic cells. Oncotarget 2018; 9:28294-28308. [PMID: 29983861 PMCID: PMC6033361 DOI: 10.18632/oncotarget.25511] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 05/13/2018] [Indexed: 12/21/2022] Open
Abstract
Melanoma is the most dangerous form of skin cancer with a growing incidence over the last decades. Fourty percent of all melanomas harbor a mutation in the signaling adaptor BRAF (V600E) that results in ERK hyperactivity as an oncogenic driver. In these cases, treatment with the BRAFV600E inhibitors Vemurafenib (VEM) or Dabrafenib (DAB) coapplied with the MEK1/2 inhibitors Cobimetinib (COB) or Trametinib (TRA) can result in long-term suppression of tumor growth. Besides direct suppression of ERK activity, these inhibitors have been reported to also modulate tumor immune responses, and exert pro-inflammatory side effects such as fever and rash in some patients. Here we asked for potential effects of BRAFV600E inhibitors on dendritic cells (DC) which are essential for the induction of adaptive anti-tumor responses. Both splenic and bone marrow-derived (BM) mouse dendritic cells (DC) up-regulated costimulator expression (CD80, CD86) in response to DAB but not VEM treatment. Moreover, DAB and to lesser extent VEM enhanced IL-1β (interleukin 1 beta) release by splenic DC, and by LPS-stimulated BMDC. We demonstrate that DAB and VEM activated the NLRC4/Caspase-1 inflammasome. At high concentration, DAB also induced inflammasome activation independent of Caspase-1. TRA and COB elevated MHCII expression on BMDC, and modulated the LPS-induced cytokine pattern. Immunomodulatory activity of DAB and VEM was also observed in human monocyte-derived DC, and DAB induced IL-1β in human primary DC. Altogether, our study shows that BRAFV600E inhibitors upregulate IL-1β release by mouse and human DC which may affect the DC-mediated course of anti-tumor immune responses.
Collapse
Affiliation(s)
- Eva Hajek
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Franziska Krebs
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Rebekka Bent
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Katharina Haas
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Antje Bast
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Ivo Steinmetz
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
43
|
Zappasodi R, Merghoub T, Wolchok JD. Emerging Concepts for Immune Checkpoint Blockade-Based Combination Therapies. Cancer Cell 2018; 33:581-598. [PMID: 29634946 PMCID: PMC5896787 DOI: 10.1016/j.ccell.2018.03.005] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/13/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022]
Abstract
Checkpoint blockade has formally demonstrated that reactivating anti-tumor immune responses can regress tumors. However, this only occurs in a fraction of patients. Incorporating these therapies in more powerful combinations is thus a logical next step. Here, we review functional roles of immune checkpoints and molecular determinants of checkpoint-blockade clinical activity. Limited-size T cell-infiltrated tumors, differing substantially from "self," generally respond to checkpoint blockade. Therefore, we propose that reducing tumor burden and increasing tumor immunogenicity are key factors to improve immunotherapy. Lastly, we outline criteria to select proper immunotherapy combination partners and highlight the importance of activity biomarkers for timely treatment optimization.
Collapse
Affiliation(s)
- Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
44
|
Hargadon KM. Strategies to Improve the Efficacy of Dendritic Cell-Based Immunotherapy for Melanoma. Front Immunol 2017; 8:1594. [PMID: 29209327 PMCID: PMC5702020 DOI: 10.3389/fimmu.2017.01594] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022] Open
Abstract
Melanoma is a highly aggressive form of skin cancer that frequently metastasizes to vital organs, where it is often difficult to treat with traditional therapies such as surgery and radiation. In such cases of metastatic disease, immunotherapy has emerged in recent years as an exciting treatment option for melanoma patients. Despite unprecedented successes with immune therapy in the clinic, many patients still experience disease relapse, and others fail to respond at all, thus highlighting the need to better understand factors that influence the efficacy of antitumor immune responses. At the heart of antitumor immunity are dendritic cells (DCs), an innate population of cells that function as critical regulators of immune tolerance and activation. As such, DCs have the potential to serve as important targets and delivery agents of cancer immunotherapies. Even immunotherapies that do not directly target or employ DCs, such as checkpoint blockade therapy and adoptive cell transfer therapy, are likely to rely on DCs that shape the quality of therapy-associated antitumor immunity. Therefore, understanding factors that regulate the function of tumor-associated DCs is critical for optimizing both current and future immunotherapeutic strategies for treating melanoma. To this end, this review focuses on advances in our understanding of DC function in the context of melanoma, with particular emphasis on (1) the role of immunogenic cell death in eliciting tumor-associated DC activation, (2) immunosuppression of DC function by melanoma-associated factors in the tumor microenvironment, (3) metabolic constraints on the activation of tumor-associated DCs, and (4) the role of the microbiome in shaping the immunogenicity of DCs and the overall quality of anti-melanoma immune responses they mediate. Additionally, this review highlights novel DC-based immunotherapies for melanoma that are emerging from recent progress in each of these areas of investigation, and it discusses current issues and questions that will need to be addressed in future studies aimed at optimizing the function of melanoma-associated DCs and the antitumor immune responses they direct against this cancer.
Collapse
Affiliation(s)
- Kristian M. Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, United States
| |
Collapse
|
45
|
Passarelli A, Mannavola F, Stucci LS, Tucci M, Silvestris F. Immune system and melanoma biology: a balance between immunosurveillance and immune escape. Oncotarget 2017; 8:106132-106142. [PMID: 29285320 PMCID: PMC5739707 DOI: 10.18632/oncotarget.22190] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/21/2017] [Indexed: 12/28/2022] Open
Abstract
Melanoma is one of the most immunogenic tumors and its relationship with host immune system is currently under investigation. Many immunomodulatory mechanisms, favoring melanomagenesis and progression, have been described to interfere with the disablement of melanoma recognition and attack by immune cells resulting in immune resistance and immunosuppression. This knowledge produced therapeutic advantages, such as immunotherapy, aiming to overcome the immune evasion. Here, we review the current advances in cancer immunoediting and focus on melanoma immunology, which involves a dynamic interplay between melanoma and immune system, as well as on effects of “targeted therapies” on tumor microenvironment for combination strategies.
Collapse
Affiliation(s)
- Anna Passarelli
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Francesco Mannavola
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Luigia Stefania Stucci
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Marco Tucci
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Francesco Silvestris
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| |
Collapse
|
46
|
BRAF V600E accelerates disease progression and enhances immune suppression in a mouse model of B-cell leukemia. Blood Adv 2017; 1:2147-2160. [PMID: 29296862 DOI: 10.1182/bloodadvances.2017006593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/25/2017] [Indexed: 01/21/2023] Open
Abstract
Mutated mitogen-activated protein kinase (MAPK) pathway components promote tumor survival, proliferation, and immune evasion in solid tumors. MAPK mutations occur in hematologic cancers as well, but their role is less clear and few models are available to study this. We developed an in vivo model of disseminated BRAFV600E B-cell leukemia to determine the effects of this mutation on tumor development and immune evasion. Mice with B-cell-restricted BRAFV600E expression crossed with the Eµ-TCL1 model of chronic lymphocytic leukemia (CLL) developed leukemia significantly earlier (median, 4.9 vs 8.1 months; P < .001) and had significantly shorter lifespan (median, 7.3 vs 12.1 months; P < .001) versus BRAF wild-type counterparts. BRAFV600E expression did not affect B-cell proliferation but reduced spontaneous apoptosis. BRAFV600E-mutant leukemia produced greater T-cell effects, evidenced by exhaustion immunophenotype and CD44+ T-cell percentage, as well as increased expression of PD-L1 on CD11b+ cells. Results were confirmed in syngeneic mice engrafted with BRAFV600E leukemia cells. Furthermore, a BRAFV600E-expressing CLL cell line more strongly inhibited anti-CD3/CD28-induced T-cell proliferation, which was reversed by BRAFV600E inhibition. These results demonstrate the immune-suppressive impact of BRAFV600E in B-cell leukemias and introduce a new model to develop rational combination strategies targeting both tumor cells and tumor-mediated immune evasion.
Collapse
|
47
|
Griffin M, Scotto D, Josephs DH, Mele S, Crescioli S, Bax HJ, Pellizzari G, Wynne MD, Nakamura M, Hoffmann RM, Ilieva KM, Cheung A, Spicer JF, Papa S, Lacy KE, Karagiannis SN. BRAF inhibitors: resistance and the promise of combination treatments for melanoma. Oncotarget 2017; 8:78174-78192. [PMID: 29100459 PMCID: PMC5652848 DOI: 10.18632/oncotarget.19836] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022] Open
Abstract
Identification of mutations in the gene encoding the serine/threonine-protein kinase, BRAF, and constitutive activation of the mitogen-activated protein kinase (MAPK) pathway in around 50% of malignant melanomas have led to the development and regulatory approval of targeted pathway inhibitor drugs. A proportion of patients are intrinsically resistant to BRAF inhibitors, and most patients who initially respond, acquire resistance within months. In this review, we discuss pathway inhibitors and their mechanisms of resistance, and we focus on numerous efforts to improve clinical benefits through combining agents with disparate modes of action, including combinations with checkpoint inhibitor antibodies. We discuss the merits of combination strategies based on enhancing immune responses or overcoming tumor-associated immune escape mechanisms. Emerging insights into mechanisms of action, resistance pathways and their impact on host-tumor relationships will inform the design of optimal combinations therapies to improve outcomes for patients who currently do not benefit from recent treatment breakthroughs.
Collapse
Affiliation(s)
- Merope Griffin
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Daniele Scotto
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Debra H. Josephs
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
- Research Oncology, School of Cancer Sciences, King's College London, Guy's Hospital, Bermondsey Wing, London, UK
| | - Silvia Mele
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Silvia Crescioli
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Heather J. Bax
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
- Research Oncology, School of Cancer Sciences, King's College London, Guy's Hospital, Bermondsey Wing, London, UK
| | - Giulia Pellizzari
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
- Research Oncology, School of Cancer Sciences, King's College London, Guy's Hospital, Bermondsey Wing, London, UK
| | - Matthew D. Wynne
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Mano Nakamura
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Ricarda M. Hoffmann
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Kristina M. Ilieva
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
- Breast Cancer Now Unit, School of Cancer Sciences, King's College London, Guy's Cancer Centre, London, UK
| | - Anthony Cheung
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
- Breast Cancer Now Unit, School of Cancer Sciences, King's College London, Guy's Cancer Centre, London, UK
| | - James F. Spicer
- Research Oncology, School of Cancer Sciences, King's College London, Guy's Hospital, Bermondsey Wing, London, UK
| | - Sophie Papa
- Research Oncology, School of Cancer Sciences, King's College London, Guy's Hospital, Bermondsey Wing, London, UK
| | - Katie E. Lacy
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
| | - Sophia N. Karagiannis
- St John's Institute of Dermatology, Genetics and Molecular Medicine, King's College London, Guy's Hospital, Tower Wing, London, UK
- Breast Cancer Now Unit, School of Cancer Sciences, King's College London, Guy's Cancer Centre, London, UK
| |
Collapse
|
48
|
Liu X, Zhou Q, Xu Y, Chen M, Zhao J, Wang M. Harness the synergy between targeted therapy and immunotherapy: what have we learned and where are we headed? Oncotarget 2017; 8:86969-86984. [PMID: 29156850 PMCID: PMC5689740 DOI: 10.18632/oncotarget.21160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/05/2017] [Indexed: 12/22/2022] Open
Abstract
Since the introduction of imatinib for the treatment of chronic myelogenous leukemia, several oncogenic mutations have been identified in various malignancies that can serve as targets for therapy. More recently, a deeper insight into the mechanism of antitumor immunity and tumor immunoevasion have facilitated the development of novel immunotherapy agents. Certain targeted agents have the ability of inhibiting tumor growth without causing severe lymphocytopenia and amplifying antitumor immune response by increasing tumor antigenicity, enhancing intratumoral T cell infiltration, and altering the tumor immune microenvironment, which provides a rationale for combining targeted therapy with immunotherapy. Targeted therapy can elicit dramatic responses in selected patients by interfering with the tumor-intrinsic driver mutations. But in most cases, resistance will occur over a relatively short period of time. In contrast, immunotherapy can yield durable, albeit generally mild, responses in several tumor types via unleashing host antitumor immunity. Thus, combination approaches might be able to induce a rapid tumor regression and a prolonged duration of response. We examine the available evidence regarding immune effects of targeted therapy, and review preclinical and clinical studies on the combination of targeted therapy and immunotherapy for cancer treatment. Furthermore, we discuss challenges of the combined therapy and highlight the need for continued translational research.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qing Zhou
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yan Xu
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Minjiang Chen
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jing Zhao
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Mengzhao Wang
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
49
|
Reddy SM, Reuben A, Wargo JA. Influences of BRAF Inhibitors on the Immune Microenvironment and the Rationale for Combined Molecular and Immune Targeted Therapy. Curr Oncol Rep 2017; 18:42. [PMID: 27215436 DOI: 10.1007/s11912-016-0531-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The identification of key driver mutations in melanoma has led to the development of targeted therapies aimed at BRAF and MEK, but responses are often limited in duration. There is growing evidence that MAPK pathway activation impairs antitumor immunity and that targeting this pathway may enhance responses to immunotherapies. There is also evidence that immune mechanisms of resistance to targeted therapy exist, providing the rationale for combining targeted therapy with immunotherapy. Preclinical studies have demonstrated synergy in combining these strategies, and combination clinical trials are ongoing. It is, however, becoming clear that additional translational studies are needed to better understand toxicity, proper timing, and sequence of therapy, as well as the utility of multidrug regimens and effects of other targeted agents on antitumor immunity. Insights gained through translational research in preclinical models and clinical studies will provide mechanistic insight into therapeutic response and resistance and help devise rational strategies to enhance therapeutic responses.
Collapse
Affiliation(s)
- Sangeetha M Reddy
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Unit 463, Houston, TX, 77030, USA
| | - Alexandre Reuben
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
50
|
Lai X, Friedman A. Combination therapy for melanoma with BRAF/MEK inhibitor and immune checkpoint inhibitor: a mathematical model. BMC SYSTEMS BIOLOGY 2017; 11:70. [PMID: 28724377 PMCID: PMC5517842 DOI: 10.1186/s12918-017-0446-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 07/11/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND The B-raf gene is mutated in up to 66% of human malignant melanomas, and its protein product, BRAF kinase, is a key part of RAS-RAF-MEK-ERK (MAPK) pathway of cancer cell proliferation. BRAF-targeted therapy induces significant responses in the majority of patients, and the combination BRAF/MEK inhibitor enhances clinical efficacy, but the response to BRAF inhibitor and to BRAF/MEK inhibitor is short lived. On the other hand, treatment of melanoma with an immune checkpoint inhibitor, such as anti-PD-1, has lower response rate but the response is much more durable, lasting for years. For this reason, it was suggested that combination of BRAF/MEK and PD-1 inhibitors will significantly improve overall survival time. RESULTS This paper develops a mathematical model to address the question of the correlation between BRAF/MEK inhibitor and PD-1 inhibitor in melanoma therapy. The model includes dendritic and cancer cells, CD 4+ and CD 8+ T cells, MDSC cells, interleukins IL-12, IL-2, IL-6, IL-10 and TGF- β, PD-1 and PD-L1, and the two drugs: BRAF/MEK inhibitor (with concentration γ B ) and PD-1 inhibitor (with concentration γ A ). The model is represented by a system of partial differential equations, and is used to develop an efficacy map for the combined concentrations (γ B ,γ A ). It is shown that the two drugs are positively correlated if γ B and γ A are at low doses, that is, the growth of the tumor volume decreases if either γ B or γ A is increased. On the other hand, the two drugs are antagonistic at some high doses, that is, there are zones of (γ B ,γ A ) where an increase in one of the two drugs will increase the tumor volume growth, rather than decrease it. CONCLUSIONS It will be important to identify, by animal experiments or by early clinical trials, the zones of (γ B ,γ A ) where antagonism occurs, in order to avoid these zones in more advanced clinical trials.
Collapse
Affiliation(s)
- Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, Beijing, 100872 People’s Republic of China
| | - Avner Friedman
- Mathematical Bioscience Institute & Department of Mathematics, Ohio State University, Columbus, 43210 OH USA
| |
Collapse
|