1
|
Wu J, Ding Q, Zhang Q, Chen Q, Wen X, Ding Y, Li J, Chen Z, Zhang T, Wang J, Huang F, Jiang H, Chen L, Zhou Q, Li K, Zhang X, Li D. Addition of anti-PD-1 immunotherapy to BRAF inhibitor-based targeted therapy improves real-world survival and delays brain metastases in patients with BRAF V600-mutant advanced melanoma: a multicenter cohort study. MedComm (Beijing) 2025; 6:e70102. [PMID: 39968494 PMCID: PMC11832434 DOI: 10.1002/mco2.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 02/20/2025] Open
Abstract
Anti-PD-1 immunotherapy and targeted therapy (TT) represent two major therapeutic modalities for BRAFV600-mutant advanced melanoma, but the efficacy of combination therapy in Asian populations remains unknown. Asian melanoma patients differ significantly from Caucasians in tissue subtypes, pathogenesis and response to treatment. We retrospectively analyzed data of BRAFV600-mutant advanced melanoma patients treated with first-line vemurafenib (V) ± anti-PD-1 or dabrafenib+trametinib (D+T) ± anti-PD-1 between 2014 and 2023 from three centers in China. 178 patients were included, with V (n = 45), D+T (n = 51), V+anti-PD-1 (n = 39) and D+T+anti-PD-1 (n = 43). The median PFS (21.9 vs. 11.1 months, p < 0.001), OS (NR vs. 32.6 months, p = 0.027), and DoR (20.0 vs. 8.4 months, p = 0.002) were significantly prolonged with D+T+anti-PD-1 versus D+T. Addition of anti-PD-1 to V also significantly prolonged PFS, OS, and DoR (p < 0.001). V+anti-PD-1 was superior to D+T in terms of PFS (15.0 vs. 11.1 months, p = 0.007) and DoR (18.0 vs. 8.4 months, p = 0.013), and was comparable to D+T+anti-PD-1. Addition of anti-PD-1 to BRAF inhibitor-based TT was associated with lower incidence of brain metastases (p = 0.032). Addition of anti-PD-1 to BRAF inhibitor-based TT appears to be a safe and effective treatment option, conferring a survival benefit and delaying the onset brain metastases in patients with BRAFV600-mutant advanced melanoma.
Collapse
Affiliation(s)
- Junwan Wu
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Qiuyue Ding
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Qiong Zhang
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Qianqi Chen
- Department of OncologyHuazhong University of Science and Technology Union Shenzhen HospitalShenzhenGuangdong ProvinceP. R. China
| | - Xizhi Wen
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Ya Ding
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Jingjing Li
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Ziluan Chen
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Tao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Jiuhong Wang
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Fuxue Huang
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Hang Jiang
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Linbin Chen
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Qiming Zhou
- Department of OncologyHuazhong University of Science and Technology Union Shenzhen HospitalShenzhenGuangdong ProvinceP. R. China
| | - Ke Li
- Department of Cancer Biotherapy CenterYunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan ProvinceKunmingYunnan ProvinceP. R. China
| | - Xiaoshi Zhang
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| | - Dandan Li
- Biotherapy Center, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdong ProvinceP. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐sen University Cancer CenterGuangzhouGuangdong ProvinceP. R. China
| |
Collapse
|
2
|
Martin-Liberal J, Márquez-Rodas I, Cerezuela-Fuentes P, Soria A, Garicano F, Medina J, García Galindo R, Oramas J, Luis Manzano J, Delgado M, Valdivia J, Sanchez P. Challenges and perspectives in the management of BRAF-mutated metastatic melanoma: Systemic treatment sequencing and brain metastases. Cancer Treat Rev 2025; 133:102886. [PMID: 39879863 DOI: 10.1016/j.ctrv.2025.102886] [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: 11/06/2024] [Revised: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
The global incidence of metastatic melanoma with BRAF mutations, characterized by aggressive behavior and poor prognosis, is rising. Recent treatment advances, including immune checkpoint inhibitors (ICI) and targeted therapies (TT) such as BRAF and MEK inhibitors, have significantly enhanced patient outcomes. Although guidelines recommend sequencing strategies, real-world implementation can be influenced by clinical scenarios. This article highlights the importance of tailored treatment strategies, emphasizing that the decision to initiate immunotherapy (IT) or TT hinges on multiple factors, including tumor burden, LDH levels, presence of brain metastases, and patient symptomatic status. Managing brain metastases also poses a challenge, as these patients are typically excluded from pivotal clinical trials. While insights from phase II studies provide some guidance, there is a critical need for more quality data to inform comprehensive recommendations. Furthermore, although triple therapy combining IT and TT was initially thought to be promising, it has failed to clearly demonstrate benefit over current treatments. For all these reasons, there is an imperative need for further research on biomarkers and predictive factors, as well as real-world studies, that will help tailor treatment strategies across diverse patient subsets, thereby refining therapeutic approaches for BRAF-mutated metastatic melanoma.
Collapse
Affiliation(s)
| | - Iván Márquez-Rodas
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | | | - Ainara Soria
- Ramón y Cajal University Hospital, Madrid, Spain
| | | | - Javier Medina
- General University Hospital of Toledo, Toledo, Spain
| | | | - Juana Oramas
- University Hospital of the Canary Islands, Tenerife, Spain
| | | | - Mayte Delgado
- San Cecilio Clinical University Hospital, Granada, Spain
| | - Javier Valdivia
- Medical Oncology, Virgen de las Nieves University Hospital, Granada, Spain
| | | |
Collapse
|
3
|
de Oliveira GAF, de Oliveira DA, Nisimoto MYSM, Rubinho R, Gomes HC, Marti LC, de Oliveira Filho RS. Advances in Tumor-Infiltrating Lymphocyte (TIL) as a Prognostic Factor and for Treating Invasive Cutaneous Melanoma. Int J Mol Sci 2024; 25:12596. [PMID: 39684307 PMCID: PMC11641307 DOI: 10.3390/ijms252312596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Invasive cutaneous melanoma is responsible for about 5% of skin tumors yet is liable for nearly 70% of skin cancer-related deaths. Despite notable advancements over the past decade, including immunotherapies and targeted treatments, more than half of invasive melanoma patients ultimately succumb to the disease due to therapeutic resistance. To overcome this obstacle, strategies such as combining immunotherapies with targeted drugs or adding epigenetic therapies have been investigated. Tumor-infiltrating lymphocytes (TILs) therapy has emerged as a promising option for patients whose disease continues to progress despite standard treatments. This article aims to introduce TIL therapy and review recent outcomes in melanoma prognosis in its application for melanoma management.
Collapse
Affiliation(s)
- Gabriel Alves Freiria de Oliveira
- Melanoma and Skin Tumors Sector, Plastic Surgery Discipline, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), Rua Botucatu, 740, São Paulo 04023-062, SP, Brazil; (G.A.F.d.O.); (D.A.d.O.); (M.Y.S.M.N.); (H.C.G.)
| | - Daniel Arcuschin de Oliveira
- Melanoma and Skin Tumors Sector, Plastic Surgery Discipline, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), Rua Botucatu, 740, São Paulo 04023-062, SP, Brazil; (G.A.F.d.O.); (D.A.d.O.); (M.Y.S.M.N.); (H.C.G.)
| | - Melissa Yoshimi Sakamoto Maeda Nisimoto
- Melanoma and Skin Tumors Sector, Plastic Surgery Discipline, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), Rua Botucatu, 740, São Paulo 04023-062, SP, Brazil; (G.A.F.d.O.); (D.A.d.O.); (M.Y.S.M.N.); (H.C.G.)
| | - Rafael Rubinho
- Department of Dermatology, Universidade Santo Amaro, Rua Isabel Schmidt, 349, Santo Amaro, São Paulo 04743-030, SP, Brazil;
| | - Heitor Carvalho Gomes
- Melanoma and Skin Tumors Sector, Plastic Surgery Discipline, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), Rua Botucatu, 740, São Paulo 04023-062, SP, Brazil; (G.A.F.d.O.); (D.A.d.O.); (M.Y.S.M.N.); (H.C.G.)
| | - Luciana Cavalheiro Marti
- Department of Experimental Research, Hospital Israelita Albert Einstein, Rua Comendador Elias Jafet, 755, Morumbi, São Paulo 05653-000, SP, Brazil
| | - Renato Santos de Oliveira Filho
- Melanoma and Skin Tumors Sector, Plastic Surgery Discipline, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), Rua Botucatu, 740, São Paulo 04023-062, SP, Brazil; (G.A.F.d.O.); (D.A.d.O.); (M.Y.S.M.N.); (H.C.G.)
| |
Collapse
|
4
|
Grützner C, Tan YP, Müller P, Schlee-Guimaraes TM, Jentzsch M, Schmid-Burgk JL, Renn M, Behrendt R, Hartmann G. Synthetic RIG-I agonist-mediated cancer immunotherapy synergizes with MAP kinase inhibition against BRAF-mutated melanoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102283. [PMID: 39165562 PMCID: PMC11334831 DOI: 10.1016/j.omtn.2024.102283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024]
Abstract
The implementation of targeted molecular therapies and immunotherapy in melanoma vastly improved the therapeutic outcome in patients with limited efficacy of surgical intervention. Nevertheless, a large fraction of patients with melanoma still remain refractory or acquire resistance to these new forms of treatment, illustrating a need for improvement. Here, we report that the clinically relevant combination of mitogen-activated protein (MAP) kinase pathway inhibitors dabrafenib and trametinib synergize with RIG-I agonist-induced immunotherapy to kill BRAF-mutated human and mouse melanoma cells. Kinase inhibition did not compromise the agonist-induced innate immune response of the RIG-I pathway in host immune cells. In a melanoma transplantation mouse model, the triple therapy outperformed individual therapies. Our study suggests that agonist-induced activation of RIG-I with its synthetic ligand 3pRNA could vastly improve tumor control in a substantial fraction of patients with melanoma receiving MAP kinase inhibitors.
Collapse
Affiliation(s)
- Christian Grützner
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
- Department of Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Bonn, Germany
| | - Yu Pan Tan
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Patrick Müller
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | | | - Marius Jentzsch
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Jonathan L. Schmid-Burgk
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Marcel Renn
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Rayk Behrendt
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Gunther Hartmann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
5
|
Vuaroqueaux V, Musch A, Peille AL, Kelter G, Weichert L, Metz T, Hendriks HR, Fiebig HH. High In Vitro and In Vivo Activity of BI-847325, a Dual MEK/Aurora Kinase Inhibitor, in Human Solid and Hematologic Cancer Models. CANCER RESEARCH COMMUNICATIONS 2023; 3:2170-2181. [PMID: 37830744 PMCID: PMC10599287 DOI: 10.1158/2767-9764.crc-22-0221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/23/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
BI-847325 is an ATP-competitive inhibitor of MEK/Aurora kinases with the potential to treat a wide range of cancers. In a panel of 294 human tumor cell lines in vitro, BI-847325 was found to be a highly selective inhibitor that was active in the submicromolar range. The most sensitive cancer types were acute lymphocytic and myelocytic leukemia, melanomas, bladder, colorectal, and mammary cancers. BI-847325 showed a broader range of activity than the MEK inhibitor GDC-0623. The high efficacy of BI-847325 was associated with but not limited to cell lines with oncogenic mutations in NRAS, BRAF, and MAP2K1.The high antiproliferative activity of BI-847325 was validated in vivo using subcutaneous xenograft models. After oral administration of 80 and 40 mg/kg once weekly for 3 or 4 weeks, BI-847325 was highly active in four of five colorectal, two of two gastric, two of two mammary, and one of one pancreatic cancer models (test/control < 25%), and tumor regressions were observed in five of 11 cancer models. The treatment was well tolerated with no relevant lethality or body weight changes. In combination with capecitabine, BI-847325 displayed synergism over single-agent therapies, leading to complete remission in the triple-negative mammary model MAXFTN 401, partial regression in the colon model CXF 1103, and stasis in the gastric models GXA 3011 and GXA 3023. In conclusion, dual MEK/Aurora kinase inhibition shows remarkable potential for treating multiple types of hematologic and solid tumors. The combination with capecitabine was synergistic in colorectal, gastric, and mammary cancer. SIGNIFICANCE We report the preclinical evaluation of BI-847325, a MEK/Aurora kinase inhibitor. Our data demonstrate that BI-847325 has potent antitumor activity in a broad range of human solid and hematologic cancer models in vitro and in vivo and is well tolerated in animal models. It also shows synergistic effect when combined with capecitabine. These findings provide a strong rationale for further development of BI-847325 as a potential therapeutic for patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Gerhard Kelter
- Charles River, Discovery Research Services GmbH, Freiburg, Germany
| | - Loreen Weichert
- Charles River, Discovery Research Services GmbH, Freiburg, Germany
| | | | | | | |
Collapse
|
6
|
Haynes D, Morgan EE, Chu EY. Cutaneous adverse reactions resulting from targeted cancer therapies: histopathologic and clinical findings. Hum Pathol 2023; 140:129-143. [PMID: 37146945 DOI: 10.1016/j.humpath.2023.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
Targeted cancer treatments-designed to interfere with specific molecular signals responsible for tumor survival and progression-have shown benefit over conventional chemotherapies but may lead to diverse cutaneous adverse effects. This review highlights clinically significant dermatologic toxicities and their associated histopathologic findings, resulting from various targeted cancer drugs. Case reports and series, clinical trials, reviews, and meta-analyses are included for analysis and summarized herein. Cutaneous side effects resulting from targeted cancer therapies were reported with incidences as high as 90% for certain medications, and reactions are often predictable based on mechanism(s) of action of a given drug. Common and important reaction patterns included: acneiform eruptions, neutrophilic dermatoses, hand-foot skin reaction, secondary cutaneous malignancies, and alopecia. Clinical and histopathologic recognition of these toxicities remains impactful for patient care.
Collapse
Affiliation(s)
- Dylan Haynes
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Eric E Morgan
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Emily Y Chu
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Sah VR, Jespersen H, Karlsson J, Nilsson LM, Bergqvist M, Johansson I, Carneiro A, Helgadottir H, Levin M, Ullenhag G, Ståhlberg A, Olofsson Bagge R, Nilsson JA, Ny L. Chemokine Analysis in Patients with Metastatic Uveal Melanoma Suggests a Role for CCL21 Signaling in Combined Epigenetic Therapy and Checkpoint Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2023; 3:884-895. [PMID: 37377898 PMCID: PMC10194136 DOI: 10.1158/2767-9764.crc-22-0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/01/2023] [Accepted: 04/28/2023] [Indexed: 06/29/2023]
Abstract
Purpose Patients with metastatic uveal melanoma have limited therapeutic options and high mortality rate so new treatment options are needed. Patients and Methods We previously reported that patients treated with the PD-1 inhibitor pembrolizumab and the histone deacetylase inhibitor entinostat in the PEMDAC trial, experienced clinical benefits if their tumor originated from iris or was wildtype for BAP1 tumor suppressor gene. Here we present the 2-year follow-up of the patients in the PEMDAC trial and identify additional factors that correlate with response or survival. Results Durable responses were observed in 4 patients, with additional 8 patients exhibiting a stable disease. The median overall survival was 13.7 months. Grade 3 adverse events were reported in 62% of the patients, but they were all manageable. No fatal toxicity was observed. Activity of thymidine kinase 1 in plasma was higher in patients with stable disease or who progressed on treatment, compared with those with partial response. Chemokines and cytokines were analyzed in plasma. Three chemokines were significantly different when comparing patients with and without response. One of the factors, CCL21, was higher in the plasma of responding patients before treatment initiation but decreased in the same patients upon treatment. In tumors, CCL21 was expressed in areas resembling tertiary lymphoid structures (TLS). High plasma levels of CCL21 and presence of TLS-like regions in the tumor correlated with longer survival. Conclusions This study provides insight into durable responses in the PEMDAC trial, and describes dynamic changes of chemokines and cytokines in the blood of these patients. Significance The most significant finding from the 2-year follow-up study of the PEMDAC trial was that high CCL21 levels in blood was associated with response and survival. CCL21 was also expressed in TLS-like regions and presence of these regions was associated with longer survival. These analyses of soluble and tumor markers can inform on predictive biomarkers needing validation and become hypothesis generating for experimental research.
Collapse
Affiliation(s)
- Vasu R. Sah
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Jespersen
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Joakim Karlsson
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | - Lisa M. Nilsson
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | | | - Iva Johansson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ana Carneiro
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Hildur Helgadottir
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Max Levin
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gustav Ullenhag
- Department of Radiology, Oncology and Radiation Science, Section of Oncology, Uppsala University, Uppsala, Sweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jonas A. Nilsson
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Campbell KM, Amouzgar M, Pfeiffer SM, Howes TR, Medina E, Travers M, Steiner G, Weber JS, Wolchok JD, Larkin J, Hodi FS, Boffo S, Salvador L, Tenney D, Tang T, Thompson MA, Spencer CN, Wells DK, Ribas A. Prior anti-CTLA-4 therapy impacts molecular characteristics associated with anti-PD-1 response in advanced melanoma. Cancer Cell 2023; 41:791-806.e4. [PMID: 37037616 PMCID: PMC10187051 DOI: 10.1016/j.ccell.2023.03.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/16/2023] [Accepted: 03/08/2023] [Indexed: 04/12/2023]
Abstract
Immune checkpoint inhibitors (ICIs), including CTLA-4- and PD-1-blocking antibodies, can have profound effects on tumor immune cell infiltration that have not been consistent in biopsy series reported to date. Here, we analyze seven molecular datasets of samples from patients with advanced melanoma (N = 514) treated with ICI agents to investigate clinical, genomic, and transcriptomic features of anti-PD-1 response in cutaneous melanoma. We find that prior anti-CTLA-4 therapy is associated with differences in genomic, individual gene, and gene signatures in anti-PD-1 responders. Anti-CTLA-4-experienced melanoma tumors that respond to PD-1 blockade exhibit increased tumor mutational burden, inflammatory signatures, and altered cell cycle processes compared with anti-CTLA-4-naive tumors or anti-CTLA-4-experienced, anti-PD-1-nonresponsive melanoma tumors. We report a harmonized, aggregate resource and suggest that prior CTLA-4 blockade therapy is associated with marked differences in the tumor microenvironment that impact the predictive features of PD-1 blockade therapy response.
Collapse
Affiliation(s)
- Katie M Campbell
- Division of Hematology/Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Meelad Amouzgar
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | | | - Timothy R Howes
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Egmidio Medina
- Division of Hematology/Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Travers
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Gabriela Steiner
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Jeffrey S Weber
- Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Jedd D Wolchok
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA; Human Oncology and Pathogenesis Program, 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
| | - James Larkin
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Silvia Boffo
- Bristol Myers Squibb Corp., Princeton, NJ 08540, USA
| | - Lisa Salvador
- Bristol Myers Squibb Corp., Princeton, NJ 08540, USA
| | - Daniel Tenney
- Bristol Myers Squibb Corp., Princeton, NJ 08540, USA
| | - Tracy Tang
- Bristol Myers Squibb Corp., Princeton, NJ 08540, USA
| | | | | | - Daniel K Wells
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Antoni Ribas
- Division of Hematology/Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90024, USA.
| |
Collapse
|
9
|
Shalata W, Steckbeck R, Polishchuk I, Cohen AY, Rouvinov K, Tokar M, Abu Jama A, Abu Saleh O, Sheva K, Yakobson A. Safety and rapid response of dabrafenib and trametinib therapy during hyperbilirubinemia in metastatic melanoma. Front Oncol 2023; 13:1102330. [PMID: 36865793 PMCID: PMC9971949 DOI: 10.3389/fonc.2023.1102330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023] Open
Abstract
This case report describes the occurrence of hyperbilirubinemia as a complication of metastatic melanoma. A 72-year-old male patient was diagnosed with BRAF V600E-mutated melanoma with metastases in the liver, lymph nodes, lungs, pancreas, and stomach. Due to a lack of clinical data and specific guidelines for the treatment of mutated metastatic melanoma patients with hyperbilirubinemia, a conference of specialists debated between initiating treatment or providing supportive care. Ultimately, the patient was started on the combination therapy of dabrafenib and trametinib. This treatment resulted in a significant therapeutic response via normalization of bilirubin levels and an impressive radiological response of metastases just one month post-treatment initiation.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center and Ben Gurion University, Beer Sheva, Israel,*Correspondence: Walid Shalata,
| | - Rachel Steckbeck
- Medical School for International Health, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Ilya Polishchuk
- Internal Medicine Ward, Soroka Medical Center & Ben-Gurion University, Beer-Sheva, Israel
| | - Ahron Yehonatan Cohen
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center and Ben Gurion University, Beer Sheva, Israel
| | - Keren Rouvinov
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center and Ben Gurion University, Beer Sheva, Israel
| | - Margarita Tokar
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center and Ben Gurion University, Beer Sheva, Israel
| | - Ashraf Abu Jama
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center and Ben Gurion University, Beer Sheva, Israel
| | - Omar Abu Saleh
- Internal Medicine Ward, Soroka Medical Center & Ben-Gurion University, Beer-Sheva, Israel,Department of Dermatology and Venereology, Emek Medical Centre, Afula, Israel
| | - Kim Sheva
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center and Ben Gurion University, Beer Sheva, Israel
| | - Alexander Yakobson
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center and Ben Gurion University, Beer Sheva, Israel
| |
Collapse
|
10
|
Tan Y, Cui A, Qian L, Li C, Wu Z, Yang Y, Han P, Huang X, Diao L. Population pharmacokinetics of FCN-159, a MEK1/2 inhibitor, in adult patients with advanced melanoma and neurofibromatosis type 1 (NF1) and model informed dosing recommendations for NF1 pediatrics. Front Pharmacol 2023; 14:1101991. [PMID: 36755948 PMCID: PMC9899833 DOI: 10.3389/fphar.2023.1101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Objective: FCN-159 is a highly active mitogen-activated extracellular signal-regulated kinase 1/2 (MEK1/2) inhibitor in patients with advanced melanoma and neurofibromatosis type 1 (NF1). We report a population pharmacokinetic (PopPK) model-based analysis of FCN-159 and its application to inform dose selection for NF1 pediatric trials. Methods: PK data collected from patients with advanced melanoma and NF1 in two clinical studies (NCT03932253 and NCT04954001) were analyzed using a non-linear mixed effects model. The adult model was adapted by incorporating allometric scaling for PK projection in 2-17 years old children. Pediatric exposure in different body surface area (BSA) bins was simulated to identify nominal doses (i.e., dose amounts given as integers) and BSA bin cutoffs to achieve exposure comparable to adults' optimal exposure across the entire pediatric BSA range. Results: The final dataset consisted of 45 subjects with a total of 1030 PK samples. The PK of FCN-159 was well-described by a 2-compartment model with first-order linear elimination and delayed first-order absorption. Covariates, including BSA, age, sex, albumin, total protein, and cancer type, were identified as statistically significant predictors of FCN-159 disposition. Simulations based on the final model projected daily doses of 4 mg/m2 QD with optimized BSA bin cutoffs would allow fixed nominal doses within each bin and result in steady state exposure approximating the adult exposure observed at the recommended phase 2 dose (RP2D) in NF1, which is 8 mg QD. Conclusion: The developed population PK model adequately described the PK profile of FCN-159, which was adapted using allometric scaling to inform dose selection for NF1 pediatric trials.
Collapse
Affiliation(s)
- Yan Tan
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd., Shanghai, China
| | - Ailing Cui
- dMed Biopharmaceutical Co., Ltd., Shanghai, China
| | - Lixuan Qian
- dMed Biopharmaceutical Co., Ltd., Shanghai, China
| | - Chao Li
- Fosun Pharma USA Inc., Princeton, MA, United States
| | - Zhuli Wu
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd., Shanghai, China
| | - Yuchen Yang
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd., Shanghai, China
| | - Pu Han
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd., Shanghai, China
| | - Xin Huang
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd., Shanghai, China
| | - Lei Diao
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd., Shanghai, China
| |
Collapse
|
11
|
Ascierto PA, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, Pereira RP, Eigentler T, Rutkowski P, Demidov L, Zhukova N, Schachter J, Yan Y, Caro I, Hertig C, Xue C, Kusters L, McArthur GA, Gutzmer R. Overall survival with first-line atezolizumab in combination with vemurafenib and cobimetinib in BRAF V600 mutation-positive advanced melanoma (IMspire150): second interim analysis of a multicentre, randomised, phase 3 study. Lancet Oncol 2023; 24:33-44. [PMID: 36460017 DOI: 10.1016/s1470-2045(22)00687-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Primary analysis of the phase 3 IMspire150 study showed improved investigator-assessed progression-free survival with first-line atezolizumab, vemurafenib, and cobimetinib (atezolizumab group) versus placebo, vemurafenib, and cobimetinib (control group) in patients with BRAFV600 mutation-positive melanoma. With a median follow-up of 18·9 months (IQR 10·4-23·8) at the primary analysis, overall survival data were immature. Here, we report the results from the second, prespecified, interim overall survival analysis. METHODS The multicentre, double-blind, placebo-controlled, randomised, phase 3 IMspire150 study was done at 108 academic and community hospitals in 20 countries. Patients aged 18 years or older with previously untreated unresectable stage IIIc or stage IV melanoma and an Eastern Cooperative Oncology Group performance status of 0 or 1 were eligible for inclusion. Patients were randomly assigned (1:1) to receive either atezolizumab (840 mg intravenously on day 1 and 15) or placebo plus vemurafenib (960 mg or 720 mg twice daily orally) and cobimetinib (60 mg once daily orally; 21 days on and 7 days off) in 28-day cycles. Atezolizumab and placebo were added to treatment regimens from cycle two onwards. Randomisation was done centrally (Durham, NC, USA) based on a permuted block randomisation scheme (block size of 4) using an interactive web-based response system and was stratified by geographical region and baseline lactate dehydrogenase concentration. Overall survival was analysed in the intention-to-treat population and safety was analysed in all patients who received at least one dose of study drug according to actual treatment received. The primary endpoint was investigator-assessed progression-free survival, which was previously reported. Here, we report the second, prespecified, interim overall survival analysis, which was planned after about 270 overall survival events had occurred. The trial is ongoing, but is no longer enrolling patients, and it is registered with ClinicalTrials.gov, NCT02908672. FINDINGS Between Jan 13, 2017, and April 26, 2018, 514 patients (median age 54 years [IQR 43-63]; 299 [58%] men and 215 [42%] women) were enrolled in the trial and randomly assigned to the atezolizumab group (256 [50%] patients) or the control group (258 [50%] patients). At the data cutoff (Sept 8, 2021), 273 patients had died (126 in the atezolizumab group and 147 in the control group). Median follow-up was 29·1 months (IQR 10·1-45·4) for the atezolizumab group versus 22·8 months (10·6-44·1) for the control group. Median overall survival was 39·0 months (95% CI 29·9-not estimable) in the atezolizumab group versus 25·8 months (22·0-34·6) in the control group (HR 0·84 [95% CI 0·66-1·06]; p=0·14). The most common adverse events of any grade in the atezolizumab group were blood creatine phosphokinase increased (123 [53%] of 231 patients), diarrhoea (116 [50%]), and pyrexia (115 [50%]). The most common adverse events of any grade in the control group were diarrhoea (157 [56%] of 280 patients), blood creatine phosphokinase increased (135 [48%]), and rash (119 [43%]). The most common grade 3-4 adverse events were increased lipase (54 [23%] of 231 patients in the atezolizumab group vs 62 [22%] of 280 patients in the control group), increased blood creatine phosphokinase (51 [22%] vs 50 [18%]), and increased alanine aminotransferase (32 [14%] vs 26 [9%]). Serious adverse events were reported in 112 (48%) patients in the atezolizumab group and 117 (42%) patients in the control group. Grade 5 adverse events were reported in eight (3%) patients in the atezolizumab group versus six (2%) patients in the control group. Two grade 5 adverse events (hepatitis fulminant and hepatic failure) in the atezolizumab group were considered to be associated with the triplet combination, and one event in the control group (pulmonary haemorrhage) was considered to be associated with cobimetinib. INTERPRETATION Additional follow-up of the IMspire150 trial showed that overall survival was not significantly improved with atezolizumab, vemurafenib, and cobimetinib compared with placebo, vemurafenib, and cobimetinib in patients with BRAFV600 mutation-positive advanced melanoma. Results of the final analysis are awaited to establish whether a significant improvement in overall survival can be achieved with long-term treatment with this triplet combination versus vemurafenib plus cobimetinib. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Pascale, Naples, Italy.
| | - Daniil Stroyakovskiy
- Healthcare Department, Moscow City Oncology Hospital Number 62 of Moscow, Moscow, Russia
| | - Helen Gogas
- First Department of Medicine, National and Kapodistian University of Athens, Athens, Greece
| | - Caroline Robert
- Department of Dermatology, Gustave Roussy Institute, Villejuif-Grand Paris, France; Department of Dermatology, Paris Saclay University, Villejuif-Grand Paris, France
| | - Karl Lewis
- Comprehensive Cancer Center, University of Colorado, Aurora, CO, USA
| | - Svetlana Protsenko
- Laboratory of Molecular Oncology, N N Petrov Institute of Oncology, St Petersburg, Russia
| | | | - Thomas Eigentler
- Department of Dermatology, Venereology, and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Piotr Rutkowski
- Department of Soft Tissue and Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Lev Demidov
- N N Blokhin Russian Cancer Research Center, Ministry of Health, Moscow, Russia
| | - Natalia Zhukova
- Department of Oncology, St Petersburg State University, St Petersburg, Russia
| | - Jacob Schachter
- Division of Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | | | - Ivor Caro
- Genentech, South San Francisco, CA, USA
| | | | - Cloris Xue
- F Hoffmann-La Roche, Mississauga, ON, Canada
| | | | - Grant A McArthur
- Melanoma and Skin Service and Cancer Biology and Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ralf Gutzmer
- Klinik für Dermatologie, Allergologie und Venerologie, Haut-Tumour-Zentrum Hannover, Medizinische Hochschule Hannover, Hannover, Germany; Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum, Minden, Germany
| |
Collapse
|
12
|
Gut Microbiota and Therapy in Metastatic Melanoma: Focus on MAPK Pathway Inhibition. Int J Mol Sci 2022; 23:ijms231911990. [PMID: 36233289 PMCID: PMC9569448 DOI: 10.3390/ijms231911990] [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: 09/13/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
Gut microbiome (GM) and its either pro-tumorigenic or anti-tumorigenic role is intriguing and constitutes an evolving landscape in translational oncology. It has been suggested that these microorganisms may be involved in carcinogenesis, cancer treatment response and resistance, as well as predisposition to adverse effects. In melanoma patients, one of the most immunogenic cancers, immune checkpoint inhibitors (ICI) and MAPK-targeted therapy—BRAF/MEK inhibitors—have revolutionized prognosis, and the study of the microbiome as a modulating factor is thus appealing. Although BRAF/MEK inhibitors constitute one of the main backbones of treatment in melanoma, little is known about their impact on GM and how this might correlate with immune re-induction. On the contrary, ICI and their relationship to GM has become an interesting field of research due to the already-known impact of immunotherapy in modulating the immune system. Immune reprogramming in the tumor microenvironment has been established as one of the main targets of microbiome, since it can induce immunosuppressive phenotypes, promote inflammatory responses or conduct anti-tumor responses. As a result, ongoing clinical trials are evaluating the role of fecal microbiota transplant (FMT), as well as the impact of using dietary supplements, antibiotics and probiotics in the prediction of response to therapy. In this review, we provide an overview of GM’s link to cancer, its relationship with the immune system and how this may impact response to treatments in melanoma patients. We also discuss insights about novel therapeutic approaches including FMT, changes in diet and use of probiotics, prebiotics and symbiotics. Finally, we hypothesize on the possible pathways through which GM may impact anti-tumor efficacy in melanoma patients treated with targeted therapy, an appealing subject of which little is known.
Collapse
|
13
|
Zhou Y, Hu F, Cui Y, Wu H, Hu S, Wei W. Bibliometric analysis of research on immunogenic cell death in cancer. Front Pharmacol 2022; 13:1029020. [PMID: 36278159 PMCID: PMC9582244 DOI: 10.3389/fphar.2022.1029020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Immunotherapy is changing the way we treat cancer. Immunogenic cell death (ICD) has received considerable attention in the treatments of various cancer types, due to the long-lasting antitumor responses elicited in human body. However, to date, no relevant bibliometric research has been reported. Methods: Publications related to ICD in cancer research were collected from the Web of Science Core Collection. Using CiteSpace, VOSviewer and an online platform, the analyses of co-author, co-citation, and co-occurrence of terms retrieved from literatures were carried out. Results: A total of 1,577 publications were included in this study. The global research literatures on ICD in cancer research have been increasing from 2005 to 2021. China, the United States and France dominated in this area and had close collaborations with many countries. Six of the top 10 most contributive institutions were from France. When it comes to author analysis, Kroemer G, Zitvogel L, Kepp O, Garg AD and Galluzzi L were in both the top 10 most productive authors and top 10 most co-cited authors lists. The co-occurring author keywords could be grouped into three clusters: “biomarkers of ICD”, “nanoparticles” and “combination therapy”. In terms of promising hotspots, keywords (author keywords and KeyWords Plus) with recent citation bursts could be summarized into two aspects: “tumor microenvironment” and “nanoparticles”. Conclusion: Increased attention has been paid to ICD in cancer treatment. However, there are still many unresolved domains in the field of ICD, such as clinical application and molecular mechanisms of this cell death process. ICD-inducing modalities combined with nanotechnology could potentiate the current immunotherapies, and will be hotspots for future research.
Collapse
Affiliation(s)
- Yan Zhou
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Fen Hu
- Department of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- Institute of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
| | - Yang Cui
- Department of Neurosurgery, Hebei Yanda Hospital, Langfang, China
| | - Haiyang Wu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Shunan Hu
- Department of Neurosurgery, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- *Correspondence: Shunan Hu, ; Wei Wei,
| | - Wei Wei
- Department of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- Institute of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, XiangYang, China
- *Correspondence: Shunan Hu, ; Wei Wei,
| |
Collapse
|
14
|
García-Gutiérrez L, Fallahi E, Aboud N, Quinn N, Matallanas D. Interaction of LATS1 with SMAC links the MST2/Hippo pathway with apoptosis in an IAP-dependent manner. Cell Death Dis 2022; 13:692. [PMID: 35941108 PMCID: PMC9360443 DOI: 10.1038/s41419-022-05147-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 01/21/2023]
Abstract
Metastatic malignant melanoma is the deadliest skin cancer, and it is characterised by its high resistance to apoptosis. The main melanoma driving mutations are part of ERK pathway, with BRAF mutations being the most frequent ones, followed by NRAS, NF1 and MEK mutations. Increasing evidence shows that the MST2/Hippo pathway is also deregulated in melanoma. While mutations are rare, MST2/Hippo pathway core proteins expression levels are often dysregulated in melanoma. The expression of the tumour suppressor RASSF1A, a bona fide activator of the MST2 pathway, is silenced by promoter methylation in over half of melanomas and correlates with poor prognosis. Here, using mass spectrometry-based interaction proteomics we identified the Second Mitochondria-derived Activator of Caspases (SMAC) as a novel LATS1 interactor. We show that RASSF1A-dependent activation of the MST2 pathway promotes LATS1-SMAC interaction and negatively regulates the antiapoptotic signal mediated by the members of the IAP family. Moreover, proteomic experiments identified a common cluster of apoptotic regulators that bind to SMAC and LATS1. Mechanistic analysis shows that the LATS1-SMAC complex promotes XIAP ubiquitination and its subsequent degradation which ultimately results in apoptosis. Importantly, we show that the oncogenic BRAFV600E mutant prevents the proapoptotic signal mediated by the LATS1-SMAC complex while treatment of melanoma cell lines with BRAF inhibitors promotes the formation of this complex, indicating that inhibition of the LATS1-SMAC might be necessary for BRAFV600E-driven melanoma. Finally, we show that LATS1-SMAC interaction is regulated by the SMAC mimetic Birinapant, which requires C-IAP1 inhibition and the degradation of XIAP, suggesting that the MST2 pathway is part of the mechanism of action of Birinapant. Overall, the current work shows that SMAC-dependent apoptosis is regulated by the LATS1 tumour suppressor and supports the idea that LATS1 is a signalling hub that regulates the crosstalk between the MST2 pathway, the apoptotic network and the ERK pathway.
Collapse
Affiliation(s)
- Lucía García-Gutiérrez
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Emma Fallahi
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Nourhan Aboud
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Niall Quinn
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
15
|
Xing J, Jia Z, Li Y, Han Y. Construction of immunotherapy-related prognostic gene signature and small molecule drug prediction for cutaneous melanoma. Front Oncol 2022; 12:939385. [PMID: 35957907 PMCID: PMC9358033 DOI: 10.3389/fonc.2022.939385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Background Cutaneous melanoma (CM), a kind of skin cancer with a high rate of advanced mortality, exhibits a wide variety of driver and transmitter gene alterations in the immunological tumor microenvironment (TME) associated with tumor cell survival and proliferation. Methods We analyzed the immunological infiltration of TME cells in normal and malignant tissues using 469 CM and 556 normal skin samples. We used a single sample gene set enrichment assay (ssGSEA) to quantify the relative abundance of 28 cells, then used the LASSO COX regression model to develop a riskScore prognostic model, followed by a small molecule drug screening and molecular docking validation, which was then validated using qRT-PCR and IHC. Results We developed a prognosis model around seven essential protective genes for the first time, dramatically elevated in tumor tissues, as did immune cell infiltration. Multivariate Cox regression results indicated that riskScore is an independent and robust prognostic indicator, and its predictive value in immunotherapy was verified. Additionally, we identified Gabapentin as a possible small molecule therapeutic for CM. Conclusions A riskScore model was developed in this work to analyze patient prognosis, TME cell infiltration features, and treatment responsiveness. The development of this model not only aids in predicting patient response to immunotherapy but also has significant implications for the development of novel immunotherapeutic agents and the promotion of tailored treatment regimens.
Collapse
Affiliation(s)
- Jiahua Xing
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Ziqi Jia
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Li
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Yan Han, ; Yan Li,
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Yan Han, ; Yan Li,
| |
Collapse
|