1
|
O'Connor MH, Rhodin KE, Tyler DS, Beasley GM. Management of In-transit Disease: Regional Therapies, Intralesional Therapies, and Systemic Therapy. Surg Oncol Clin N Am 2025; 34:393-410. [PMID: 40413006 PMCID: PMC12104569 DOI: 10.1016/j.soc.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
In-transit (IT) melanoma represents a distinct, heterogeneous pattern of disease that arises as superficial tumors along the track between the primary site and the draining regional lymph node basin. Many therapies have been explored for treatment of this disease with the goal of maximizing delivery of the therapeutic agent to the tumor while minimizing systemic toxicities. These include regional chemotherapies, intralesional injections, checkpoint inhibitors, immunomodulators, and vaccines in various combinations or as monotherapy. Here, we review the general managemnt of patients with ITmelanoma, the range of currently available treatment options, and recommendations for specific therapies for individual patients.
Collapse
Affiliation(s)
- Margaret H O'Connor
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristen E Rhodin
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Douglas S Tyler
- Department of Surgery, Texas Medical Branch, Galveston, TX, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
2
|
Soltani Rad MN, Behrouz S, Charbaghi M, Behrouz M, Zarenezhad E, Ghanbariasad A. Design, synthesis, anticancer and in silico assessment of 8-caffeinyl chalcone hybrid conjugates. RSC Adv 2024; 14:26674-26693. [PMID: 39175679 PMCID: PMC11340408 DOI: 10.1039/d4ra04787g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024] Open
Abstract
In this paper, we report the design, synthesis, and characterization of novel 8-caffeinyl chalcone hybrid conjugates, which were studied for their anticancer properties, toxicity, and in silico behavior. The synthesized compounds consist of 8-caffeinyl and chalcone structures with diverse substituents. The synthesis involved three main stages: bromination of caffeine to produce 8-BC, synthesis of chalcones, and subsequent coupling of these chalcones with 8-BC. The anticancer activity of the resulting compounds was evaluated in vitro against breast cancer MCF-7 and melanoma A-375 cell lines, revealing certain compounds to have significant efficacy compared to the reference drug methotrexate. Toxicity assessments using a healthy cell line indicated that most compounds displayed some level of toxicity, with only a few exceptions. Molecular docking studies indicated robust binding affinities of selected compounds to B-RAF kinase and hDHFR enzymes. In silico analyses of pharmacokinetic and physicochemical properties demonstrated that the majority of the compounds adhered to Lipinski's rule of five. Furthermore, density functional theory (DFT) studies were performed to gain deeper insights into the properties of the intermediates used throughout the research.
Collapse
Affiliation(s)
- Mohammad Navid Soltani Rad
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98-71-3735-4520 +98-71-3735-4500
- Medicinal Chemistry Research Laboratory, Novel Technology for Health Research Center, Shiraz University of Technology Shiraz 71555-313 Iran
| | - Somayeh Behrouz
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98-71-3735-4520 +98-71-3735-4500
- Medicinal Chemistry Research Laboratory, Novel Technology for Health Research Center, Shiraz University of Technology Shiraz 71555-313 Iran
| | - Maedeh Charbaghi
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98-71-3735-4520 +98-71-3735-4500
| | - Marzieh Behrouz
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98-71-3735-4520 +98-71-3735-4500
| | - Elham Zarenezhad
- Non-communicable Diseases Research Center, Fasa University of Medical Sciences Fasa Iran
| | - Ali Ghanbariasad
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences Fasa Iran
| |
Collapse
|
3
|
Németh A, Bányai GL, Dobos NK, Kós T, Gaál A, Varga Z, Buzás EI, Khamari D, Dank M, Takács I, Szász AM, Garay T. Extracellular vesicles promote migration despite BRAF inhibitor treatment in malignant melanoma cells. Cell Commun Signal 2024; 22:282. [PMID: 38778340 PMCID: PMC11110207 DOI: 10.1186/s12964-024-01660-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Extracellular vesicles (EVs) constitute a vital component of intercellular communication, exerting significant influence on metastasis formation and drug resistance mechanisms. Malignant melanoma (MM) is one of the deadliest forms of skin cancers, because of its high metastatic potential and often acquired resistance to oncotherapies. The prevalence of BRAF mutations in MM underscores the importance of BRAF-targeted therapies, such as vemurafenib and dabrafenib, alone or in combination with the MEK inhibitor, trametinib. This study aimed to elucidate the involvement of EVs in MM progression and ascertain whether EV-mediated metastasis promotion persists during single agent BRAF (vemurafenib, dabrafenib), or MEK (trametinib) and combined BRAF/MEK (dabrafenib/trametinib) inhibition.Using five pairs of syngeneic melanoma cell lines, we assessed the impact of EVs - isolated from their respective supernatants - on melanoma cell proliferation and migration. Cell viability and spheroid growth assays were employed to evaluate proliferation, while migration was analyzed through mean squared displacement (MSD) and total traveled distance (TTD) measurements derived from video microscopy and single-cell tracking.Our results indicate that while EV treatments had remarkable promoting effect on cell migration, they exerted only a modest effect on cell proliferation and spheroid growth. Notably, EVs demonstrated the ability to mitigate the inhibitory effects of BRAF inhibitors, albeit they were ineffective against a MEK inhibitor and the combination of BRAF/MEK inhibitors. In summary, our findings contribute to the understanding of the intricate role played by EVs in tumor progression, metastasis, and drug resistance in MM.
Collapse
Affiliation(s)
- Afrodité Németh
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Gréta L Bányai
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Nikolett K Dobos
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Tamás Kós
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Anikó Gaál
- Institute of Materials and Environmental Chemistry; Biological Nanochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Zoltán Varga
- Institute of Materials and Environmental Chemistry; Biological Nanochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- ELKH-SE Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SE Extracellular Vesicle Research Group, Budapest, Hungary
| | - Delaram Khamari
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Magdolna Dank
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - István Takács
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - A Marcell Szász
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - Tamás Garay
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
4
|
Kozyra P, Pitucha M. Revisiting the Role of B-RAF Kinase as a Therapeutic Target in Melanoma. Curr Med Chem 2024; 31:2003-2020. [PMID: 37855341 DOI: 10.2174/0109298673258495231011065225] [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: 04/18/2023] [Revised: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 10/20/2023]
Abstract
Malignant melanoma is the rarest but most aggressive and deadly skin cancer. Melanoma is the result of a malignant transformation of melanocytes, which leads to their uncontrolled proliferation. Mutations in the mitogen-activated protein kinase (MAPK) pathway, which are crucial for the control of cellular processes, such as apoptosis, division, growth, differentiation, and migration, are one of its most common causes. BRAF kinase, as one of the known targets of this pathway, has been known for many years as a prominent molecular target in melanoma therapy, and the following mini-review outlines the state-of-the-art knowledge regarding its structure, mutations and mechanisms.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL, 20093, Poland
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL-20093, Poland
| |
Collapse
|
5
|
Wang K, Xiang YJ, Yu HM, Cheng YQ, Feng JK, Liu ZH, Shan YF, Zheng YT, Ni QZ, Cheng SQ. Overall survival of patients with hepatocellular carcinoma treated with sintilimab and disease outcome after treatment discontinuation. BMC Cancer 2023; 23:1017. [PMID: 37867191 PMCID: PMC10591394 DOI: 10.1186/s12885-023-11485-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND The use of Anti-PD-1 therapy has yielded promising outcomes in hepatocellular carcinoma (HCC). However, limited research has been conducted on the overall survival (OS) of patients with varying tumor responses and treatment duration. METHODS This retrospective study analyzed HCC patients who received sintilimab between January 2019 and December 2020 at four centers in China. The evaluation of tumor progression was based on Response Evaluation Criteria in Solid Tumors version 1.1. The study investigated the correlation between tumor response and OS, and the impact of drug use on OS following progressive disease (PD). RESULTS Out of 441 treated patients, 159 patients satisfied the inclusion criteria. Among them, 77 patients with disease control exhibited a significantly longer OS compared to the 82 patients with PD (median OS 26.0 vs. 11.3 months, P < 0.001). Additionally, the OS of patients with objective response (OR) was better than that of patients with stable disease (P = 0.002). Among the 47 patients with PD who continued taking sintilimab, the OS was better than the 35 patients who discontinued treatment (median OS 11.4 vs. 6.9 months, P = 0.042). CONCLUSIONS In conclusion, the tumor response in HCC patients who received sintilimab affects OS, and patients with PD may benefit from continued use of sintilimab.
Collapse
Affiliation(s)
- Kang Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yan-Jun Xiang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Hong-Ming Yu
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yu-Qiang Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jin-Kai Feng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zong-Han Liu
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yun-Feng Shan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi-Tao Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Qian-Zhi Ni
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shu-Qun Cheng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200433, China.
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China.
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200083, China.
| |
Collapse
|
6
|
Soltani Rad MN, Behrouz S, Shahbazkhani K, Behrouz M, Zarenezhad E, Ghanbariasad A. Design, synthesis, anticancer and in silico assessment of 8-piperazinyl caffeinyl-triazolylmethyl hybrid conjugates. RSC Adv 2023; 13:24656-24673. [PMID: 37601597 PMCID: PMC10436028 DOI: 10.1039/d3ra04817a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023] Open
Abstract
In this paper, we have assessed the design, synthesis, characterization, anticancer properties, toxicity, and in silico study of 8-piperazinyl caffeinyl-triazolylmethyl derivatives as new caffeine hybrid conjugates. These compounds consist of four moieties comprising 8-caffeinyl, piperazinyl, 1,2,3-triazolyl, and alkyl substituents. The synthesis of these compounds was started by bromination of caffeine to attain 8-BC, SNAr reaction with piperazine to acquire 8-PC, N-propargylation of 8-PC and finally click Huisgen cycloaddition with diverse alkyl azides. These compounds were in vitro tested against two significant cancer cell lines comprising breast cancer MCF-7 (ATCC HTB-22) and melanoma cancer A-375 (ATCC CRL-1619) cell lines and activities compared with methotrexate (MTX) as a reference drug. Anticancer assessments indicated 12j (IC50 = 323 ± 2.6) and 12k (IC50 = 175 ± 3.2) were the most potent compounds against A-375 and MCF-7 cell growth, respectively and their activities were even stronger than MTX (IC50 = 418 ± 2 for A375 and IC50 = 343 ± 3.6 for MCF-7). Toxicities were determined by screening compounds against normal cell line HEK-293 (ATCC CRL-11268) and indicated that except 12i (IC50 = 371 ± 2.3), 12j (IC50 = 418 ± 2.4), and MTX (IC50 = 199 ± 2.4), all compounds are non-toxic. Docking was conducted for 12j and 12k and determined the strong binding affinities to B-RAF kinase and hDHFR enzymes, respectively. In silico pharmacokinetic and physiochemical profiles of tested compounds were investigated which indicated that most compounds obeyed Lipinski's rule of five (RO5). The DFT study on M06-2X/6-311G (d,p) was used to indicate HOMO, LUMO, MEP, and other parameters for a better understanding of 12j and 12k reactivity. Owing to anticancer properties, toxicity, and in silico data, 12j and 12k can be proposed for further research in the future.
Collapse
Affiliation(s)
- Mohammad Navid Soltani Rad
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
- Medicinal Chemistry Research Laboratory, Novel Technology for Health Research Center, Shiraz University of Technology Shiraz 71555-313 Iran
| | - Somayeh Behrouz
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
- Medicinal Chemistry Research Laboratory, Novel Technology for Health Research Center, Shiraz University of Technology Shiraz 71555-313 Iran
| | - Kiana Shahbazkhani
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| | - Marzieh Behrouz
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| | - Elham Zarenezhad
- Non-communicable Diseases Research Center, Fasa University of Medical Sciences Fasa Iran
| | - Ali Ghanbariasad
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences Fasa Iran
| |
Collapse
|
7
|
Jain V, Harper SL, Versace AM, Fingerman D, Brown GS, Bhardwaj M, Crissey MAS, Goldman AR, Ruthel G, Liu Q, Zivkovic A, Stark H, Herlyn M, Gimotty PA, Speicher DW, Amaravadi RK. Targeting UGCG Overcomes Resistance to Lysosomal Autophagy Inhibition. Cancer Discov 2023; 13:454-473. [PMID: 36331284 PMCID: PMC9905280 DOI: 10.1158/2159-8290.cd-22-0535] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/10/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Lysosomal autophagy inhibition (LAI) with hydroxychloroquine or DC661 can enhance cancer therapy, but tumor regrowth is common. To elucidate LAI resistance, proteomics and immunoblotting demonstrated that LAI induced lipid metabolism enzymes in multiple cancer cell lines. Lipidomics showed that LAI increased cholesterol, sphingolipids, and glycosphingolipids. These changes were associated with striking levels of GM1+ membrane microdomains (GMM) in plasma membranes and lysosomes. Inhibition of cholesterol/sphingolipid metabolism proteins enhanced LAI cytotoxicity. Targeting UDP-glucose ceramide glucosyltransferase (UGCG) synergistically augmented LAI cytotoxicity. Although UGCG inhibition decreased LAI-induced GMM and augmented cell death, UGCG overexpression led to LAI resistance. Melanoma patients with high UGCG expression had significantly shorter disease-specific survival. The FDA-approved UGCG inhibitor eliglustat combined with LAI significantly inhibited tumor growth and improved survival in syngeneic tumors and a therapy-resistant patient-derived xenograft. These findings nominate UGCG as a new cancer target, and clinical trials testing UGCG inhibition in combination with LAI are warranted. SIGNIFICANCE We discovered UGCG-dependent lipid remodeling drives resistance to LAI. Targeting UGCG with a drug approved for a lysosomal storage disorder enhanced LAI antitumor activity without toxicity. LAI and UGCG inhibition could be tested clinically in multiple cancers. This article is highlighted in the In This Issue feature, p. 247.
Collapse
Affiliation(s)
- Vaibhav Jain
- Abramson Cancer Center and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Amanda M. Versace
- Abramson Cancer Center and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | - Monika Bhardwaj
- Abramson Cancer Center and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mary Ann S. Crissey
- Abramson Cancer Center and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Gordon Ruthel
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Qin Liu
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Aleksandra Zivkovic
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225, Düsseldorf, Germany
| | - Holgar Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225, Düsseldorf, Germany
| | | | - Phyllis A. Gimotty
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David W. Speicher
- The Wistar Institute, Philadelphia, PA 19104, USA
- Corresponding authors: Ravi K. Amaravadi, MD, University of Pennsylvania, 852 BRB 2/3, 421 Curie Blvd, Philadelphia, PA 19104, Tel: 215-796-5159, ; David W. Speicher, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, Tel: 215-898-3972,
| | - Ravi K. Amaravadi
- Abramson Cancer Center and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding authors: Ravi K. Amaravadi, MD, University of Pennsylvania, 852 BRB 2/3, 421 Curie Blvd, Philadelphia, PA 19104, Tel: 215-796-5159, ; David W. Speicher, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, Tel: 215-898-3972,
| |
Collapse
|
8
|
Soltani Rad MN, Behrouz S, Aghajani S, Behrouz M, Zarenezhad E, Ghanbariasad A. Design, synthesis, anticancer and in silico assessment of 8-caffeinyl-triazolylmethoxy hybrid conjugates. RSC Adv 2023; 13:3056-3070. [PMID: 36756447 PMCID: PMC9850771 DOI: 10.1039/d2ra07683g] [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: 12/02/2022] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
In this research the synthesis, characterization, anticancer and the cytotoxicity assessments of novel 8-caffeinyl-triazolylmethoxy hybrid conjugates have been described. These compounds are the first caffeine-1,2,3-triazolyl hybrid molecules that structurally are composed of three compartments comprising caffeinyl, 1,2,3-triazolyl and N-alkyl/aryl residues. The in vitro evaluations of synthesized compounds on cancer cell lines, including two breast cancer cell lines MDA-MB-468 (ATCC HTB-22), MCF-7 (ATCC HTB-22), melanoma cell line A-375 (ATCC CRL-1619) and normal cell line HEK-293 (ATCC CRL-11268) have determined that 22c (IC50 < 12.5 μM) demonstrated potent activity against A375 and its toxicity is even stronger than methotrexate (MTX) as a standard drug. Additionally, 22c involves more selectivity than MTX regarding its non-toxicity for the HEK-293 cell line. Among the tested compounds against two breast cancer cell lines, 22f (IC50 = 136 ± 0.2 and 126 ± 0.6 μM for MCF-7 and MDA-MB-468, respectively) and 22i (IC50 = 165 ± 1.8 and 175 ± 1.4 μM for MCF-7 and MDA-MB-468, respectively) were the most potent compounds but their activities were less than MTX, moreover 22f showed more selectivity regarding its lower toxicity against HEK-293. Overall, 22f displayed general toxicity and selectivity on all tested cancer cell lines. The in silico physicochemical properties, pharmacokinetic profile, and drug likeness predictions were also carried out for all the studied compounds. Most new compounds exhibited zero violation of Lipinski's rule (RO5). A molecular docking study was also conducted to predict the binding mode and the interaction of 22c as the most active anti-melanoma entry with B-RAF V600E kinase enzyme. The docking results determined that 22c exhibited a strong binding affinity to the active site of the enzyme. These findings demonstrated 22c and 22f as potential future anticancer drug candidates.
Collapse
Affiliation(s)
- Mohammad Navid Soltani Rad
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| | - Somayeh Behrouz
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| | - Saleh Aghajani
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| | - Marzieh Behrouz
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| | - Elham Zarenezhad
- Non-communicable Diseases Research Center, Fasa University of Medical Sciences Fasa Iran
| | - Ali Ghanbariasad
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences Fasa Iran
| |
Collapse
|
9
|
Mennitto A, Zattarin E, Di Maio M, Bimbatti D, De Giorgi U, Buti S, Santini D, Casadei C, Sorarù M, Messina C, Mucciarini C, Di Lorenzo G, Roviello G, Buttigliero C, Stellato M, Sepe P, Claps M, Guadalupi V, Ottini A, Pignata S, De Braud FG, Verzoni E, Procopio G. Cabozantinib beyond progression improves survival in advanced renal cell carcinoma patients: the CABEYOND study (Meet-URO 21). Expert Rev Anticancer Ther 2022; 22:115-121. [PMID: 34738499 DOI: 10.1080/14737140.2022.2002688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/25/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Cabozantinib improves survival in metastatic renal cell carcinoma (mRCC) after prior antiangiogenics. The best treatment at disease progression (PD) is unknown. Being also a AXL/MET inhibitor, involved in acquired resistance, we hypothesized a prolonged tumor growth control in patients continuing cabozantinib despite PD. RESEARCH DESIGN AND METHODS This retrospective multicenter study enrolled patients receiving cabozantinib after the first line between 2014 and 2020. We compared patients maintaining cabozantinib after first PD due to clinical benefit and good tolerability with those who changed therapy. The postprogression survival (PPS) of both was our primary endpoint. RESULTS We analyzed 89 patients: 45 received cabozantinib beyond PD and 44 switched therapy. 40.4%, 31.5%, and 28.1% of patients received 1, 2, or >2 prior treatment, respectively. 84.3% were intermediate-poor International Metastatic Renal Cell Carcinoma Database risk. Patients continuing cabozantinib showed a higher response rate to cabozantinib before PD (46.7% vs 25%, p = 0.03) and were more heavily pretreated. Continuing cabozantinib showed a significantly longer PPS compared with switching therapy (median PPS 16.9 vs 13.2 months, HR 0.66, 95%CI 0.48-0.92, p = 0.011). CONCLUSIONS We observed longer PPS in patients continuing cabozantinib beyond PD, suggesting that this could be an effective option.
Collapse
Affiliation(s)
- Alessia Mennitto
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| | - Emma Zattarin
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, Turin, Italy
| | - Davide Bimbatti
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto Iov Irccs, Padua, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per Lo Studio E La Cura Dei Tumori (Irst), Irccs, Meldola, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Chiara Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per Lo Studio E La Cura Dei Tumori (Irst), Irccs, Meldola, Italy
| | - Mariella Sorarù
- Medical Oncology, Camposampiero Hospital, Camposampiero (Padua), Italy
| | - Carlo Messina
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | | | - Giuseppe Di Lorenzo
- Oncology Unit, Andrea Tortora Hospital, ASL Salerno, Pagani, Italy
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | | | - Marco Stellato
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Pierangela Sepe
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| | - Melanie Claps
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| | - Valentina Guadalupi
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| | - Arianna Ottini
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori Irccs Fondazione "G. Pascale", Naples, Italy
| | - Filippo G De Braud
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Elena Verzoni
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione Irccs Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
10
|
Umar AB, Uzairu A, Shallangwa GA, Uba S. Ligand-based drug design and molecular docking simulation studies of some novel anticancer compounds on MALME-3M melanoma cell line. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-020-00126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Melanoma cancer causes serious health problem worldwide because of its rapid invasion to other organs and lack of satisfactory chemotherapy. The pGI50 anticancer activity values of 70 compounds from the NCI (National Cancer Institute) on MALME-3M cell line was modeled to describe the quantitative structure-activity relationships (QSARs) of the compounds, and some selected compounds were docked.
Results
The generated QSAR model was found to be statistically significant based on the obtained values of the validation keys such as R2 (0.885), $$ {R}_{\mathrm{adjusted}}^2 $$
R
adjusted
2
(0.868), Q2cv (0.842), and $$ {R}_{pred}^2 $$
R
pred
2
(0.738) required to evaluate the strength and robustness of QSAR model. Compound 39 was selected as a template due to its good pGI50 (9.205) and was modified to design new potent compounds. The predicted pGI50 activity of the designed compounds by the built model was N1 (9.836), N2 (12.876), N3 (10.901), and N4 (11.263) respectively. These proposed compounds were docked with V600E-BRAF receptor and the result shows that, N1, N2, N3, and N4 with free binding energy (FBE) of − 11.7 kcal mol−1, − 12.8 kcal mol−1, − 12.7 kcal mol−1, and − 12.9 kcal mol−1 respectively were better than the parent structure of the template (compound 39, FBE = − 7.0 kcal mol−1) and the standard V600E-BRAF inhibitor (Vemurafenib, FBE = − 11.3 kcal mol−1). Additionally, these compounds passed the drug-likeness criteria successfully to be orally bioavailable.
Conclusion
The proposed compounds were considered optimal as their performances are comparable to vemurafenib and possessed enhanced physicochemical properties. Thus recommends further research such as synthesis, in vivo, and ex-vivo evaluation.
Collapse
|
11
|
Clinical benefit of continuing pembrolizumab treatment beyond progression in patients with metastatic urothelial carcinoma. Cancer Immunol Immunother 2021; 71:229-236. [PMID: 34100985 DOI: 10.1007/s00262-021-02980-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/02/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND There has been no clinical evidence to justify continued pembrolizumab therapy beyond progression in patients with metastatic urothelial carcinoma (UC). MATERIALS AND METHODS We conducted a multicenter retrospective study evaluating the clinical efficacy of continued use of pembrolizumab beyond progression in patients with metastatic UC. Data from 51 patients with metastatic UC, who developed progression during second-line pembrolizumab therapy, were analyzed. Progression was defined based on the Immunotherapy Response Evaluation Criteria in Solid Tumors. The outcome was overall survival (OS). The association between continued treatment, OS, and the risk of all-cause mortality was tested using log-rank test, conventional and time-dependent Cox regression models. RESULTS No significant difference in patient characteristics was noted between patients continuing pembrolizumab beyond progression (N = 21) and those discontinuing pembrolizumab (N = 30). Median OS was significantly longer in the continuation group (17.8 vs. 8.8 months; P = 0.038). A multivariable conventional Cox regression model identified continued pembrolizumab administration as a significant independent prognostic factor of all-cause mortality (hazard ratio [HR]: 0.21, 95% confidence interval [CI]: 0.05-0.90, P = 0.036), irrespective of the time from treatment initiation to progression and concurrent clinical progression. Further, longer duration of pembrolizumab treatment beyond progression was independently associated with a reduced risk of all-cause mortality in a multivariable time-dependent Cox regression model, when used as a time-dependent variable (HR: 0.07, 95% CI: 0.01-0.45, P = 0.006). CONCLUSIONS Continued pembrolizumab administration beyond progression might be beneficial in patients with metastatic UC who were clinically stable.
Collapse
|
12
|
Váraljai R, Horn S, Sucker A, Piercianek D, Schmitt V, Carpinteiro A, Becker KA, Reifenberger J, Roesch A, Felsberg J, Reifenberger G, Sure U, Schadendorf D, Helfrich I. Integrative Genomic Analyses of Patient-Matched Intracranial and Extracranial Metastases Reveal a Novel Brain-Specific Landscape of Genetic Variants in Driver Genes of Malignant Melanoma. Cancers (Basel) 2021; 13:cancers13040731. [PMID: 33578810 PMCID: PMC7916600 DOI: 10.3390/cancers13040731] [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: 01/05/2021] [Revised: 01/28/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Development of brain metastases in advanced melanoma patients is a frequent event that limits patients' quality of life and survival. Despite recent insights into melanoma genetics, systematic analyses of genetic alterations in melanoma brain metastasis formation are lacking. Moreover, whether brain metastases harbor distinct genetic alterations beyond those observed at different anatomic sites of the same patient remains unknown. EXPERIMENTAL DESIGN AND RESULTS In our study, 54 intracranial and 18 corresponding extracranial melanoma metastases were analyzed for mutations using targeted next generation sequencing of 29 recurrently mutated driver genes in melanoma. In 11 of 16 paired samples, we detected nucleotide modifications in brain metastases that were absent in matched metastases at extracranial sites. Moreover, we identified novel genetic variants in ARID1A, ARID2, SMARCA4 and BAP1, genes that have not been linked to brain metastases before; albeit most frequent mutations were found in ARID1A, ARID2 and BRAF. Conclusion: Our data provide new insights into the genetic landscape of intracranial melanoma metastases supporting a branched evolution model of metastasis formation.
Collapse
Affiliation(s)
- Renáta Váraljai
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Susanne Horn
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Faculty Rudolf-Schönheimer-Institute for Biochemistry, University of Leipzig, 04103 Leipzig, Germany
| | - Antje Sucker
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Daniela Piercianek
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Department of Neurosurgery, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany
| | - Verena Schmitt
- Institute of Anatomy, Medical Faculty, University Duisburg-Essen, 45147 Essen, Germany;
| | - Alexander Carpinteiro
- Department of Molecular Biology, Medical Faculty, University Duisburg-Essen, 45147 Essen, Germany; (A.C.); (K.A.B.)
| | - Katrin Anne Becker
- Department of Molecular Biology, Medical Faculty, University Duisburg-Essen, 45147 Essen, Germany; (A.C.); (K.A.B.)
| | - Julia Reifenberger
- Department of Dermatology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Alexander Roesch
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Jörg Felsberg
- Institute of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Guido Reifenberger
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Institute of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Ulrich Sure
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Department of Neurosurgery, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Correspondence: ; Tel.: +49-201-723-1648; Fax: +49-201-723-5525
| |
Collapse
|
13
|
Clinical Implications of Acquired BRAF Inhibitors Resistance in Melanoma. Int J Mol Sci 2020; 21:ijms21249730. [PMID: 33419275 PMCID: PMC7766699 DOI: 10.3390/ijms21249730] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Understanding the role of mitogen-activated protein kinase (MAPK) pathway-activating mutations in the development and progression of melanoma and their possible use as therapeutic targets has substantially changed the management of this neoplasm, which, until a few years ago, was burdened by severe mortality. However, the presence of numerous intrinsic and extrinsic mechanisms of resistance to BRAF inhibitors compromises the treatment responses’ effectiveness and durability. The strategy of overcoming these resistances by combination therapy has proved successful, with the additional benefit of reducing side effects derived from paradoxical activation of the MAPK pathway. Furthermore, the use of other highly specific inhibitors, intermittent dosing schedules and the association of combination therapy with immune checkpoint inhibitors are promising new therapeutic strategies. However, numerous issues related to dose, tolerability and administration sequence still need to be clarified, as is to be expected from currently ongoing trials. In this review, we describe the clinical results of using BRAF inhibitors in advanced melanoma, with a keen interest in strategies aimed at overcoming resistance.
Collapse
|
14
|
Keilholz U, Ascierto PA, Dummer R, Robert C, Lorigan P, van Akkooi A, Arance A, Blank CU, Chiarion Sileni V, Donia M, Faries MB, Gaudy-Marqueste C, Gogas H, Grob JJ, Guckenberger M, Haanen J, Hayes AJ, Hoeller C, Lebbé C, Lugowska I, Mandalà M, Márquez-Rodas I, Nathan P, Neyns B, Olofsson Bagge R, Puig S, Rutkowski P, Schilling B, Sondak VK, Tawbi H, Testori A, Michielin O. ESMO consensus conference recommendations on the management of metastatic melanoma: under the auspices of the ESMO Guidelines Committee. Ann Oncol 2020; 31:1435-1448. [PMID: 32763453 DOI: 10.1016/j.annonc.2020.07.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
Abstract
The European Society for Medical Oncology (ESMO) held a consensus conference on melanoma on 5-7 September 2019 in Amsterdam, The Netherlands. The conference included a multidisciplinary panel of 32 leading experts in the management of melanoma. The aim of the conference was to develop recommendations on topics that are not covered in detail in the current ESMO Clinical Practice Guideline and where available evidence is either limited or conflicting. The main topics identified for discussion were (i) the management of locoregional disease; (ii) targeted versus immunotherapies in the adjuvant setting; (iii) targeted versus immunotherapies for the first-line treatment of metastatic melanoma; (iv) when to stop immunotherapy or targeted therapy in the metastatic setting; and (v) systemic versus local treatment for brain metastases. The expert panel was divided into five working groups to each address questions relating to one of the five topics outlined above. Relevant scientific literature was reviewed in advance. Recommendations were developed by the working groups and then presented to the entire panel for further discussion and amendment before voting. This manuscript presents the results relating to the management of metastatic melanoma, including findings from the expert panel discussions, consensus recommendations and a summary of evidence supporting each recommendation. All participants approved the final manuscript.
Collapse
Affiliation(s)
- U Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - P A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - R Dummer
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - C Robert
- Department of Dermatology, Gustave Roussy, Villejuif, France; Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - P Lorigan
- Division of Cancer Sciences, The University of Manchester and The Christie NHS Foundation Trust, Manchester, UK
| | - A van Akkooi
- Department of Surgical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - A Arance
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - C U Blank
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - V Chiarion Sileni
- Department of Experimental and Clinical Oncology, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy
| | - M Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark; University of Copenhagen, Copenhagen, Denmark
| | - M B Faries
- Department of Surgery, The Angeles Clinic, Cedars Sinai Medical Center, Los Angeles, USA
| | - C Gaudy-Marqueste
- Department of Dermatology and Skin Cancer, Aix Marseille University, Hôpital De La Timone, Marseille, France
| | - H Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - J J Grob
- Department of Dermatology and Skin Cancer, Aix Marseille University, Hôpital De La Timone, Marseille, France
| | - M Guckenberger
- Department of Radio-Oncology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - J Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A J Hayes
- Department of Academic Surgery, Royal Marsden NHS Foundation Trust, London, UK
| | - C Hoeller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - C Lebbé
- AP-HP Dermatology, Université de Paris, Paris, France; INSERM U976, Hôpital Saint Louis, Paris, France
| | - I Lugowska
- Early Phase Clinical Trials Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - M Mandalà
- Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | - I Márquez-Rodas
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - P Nathan
- Department of Medical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - B Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - R Olofsson Bagge
- Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Region Västra Götaland, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden
| | - S Puig
- Dermatology Service, Hospital Clínic of Barcelona and University of Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; CIBER, Instituto de Salud Carlos III, Barcelona, Spain
| | - P Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - B Schilling
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - V K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, USA
| | - H Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A Testori
- Department of Dermatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - O Michielin
- Department of Oncology, University Hospital Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Becco P, Gallo S, Poletto S, Frascione MPM, Crotto L, Zaccagna A, Paruzzo L, Caravelli D, Carnevale-Schianca F, Aglietta M. Melanoma Brain Metastases in the Era of Target Therapies: An Overview. Cancers (Basel) 2020; 12:cancers12061640. [PMID: 32575838 PMCID: PMC7352598 DOI: 10.3390/cancers12061640] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
Malignant melanoma is the third most common type of tumor that causes brain metastases. Patients with cerebral involvement have a dismal prognosis and their treatment is an unmet medical need. Brain involvement is a multistep process involving several signaling pathways such as Janus kinase/signal Transducer and Activator of Transcription (JAK/STAT), Phosphoinositide 3-kinase/Protein Kinase B (PI3K/AKT), Vascular Endothelial Growth Factor and Phosphatase and Tensin Homolog (PTEN). Recently therapy that targets the MAPK signaling (BRAF/MEK inhibitors) and immunotherapy (anti-CTLA4 and anti-PD1 agents) have changed the therapeutic approaches to stage IV melanoma. In contrast, there are no solid data about patients with brain metastases, who are usually excluded from clinical trials. Retrospective data showed that BRAF-inhibitors, alone or in combination with MEK-inhibitors have interesting clinical activity in this setting. Prospective data about the combinations of BRAF/MEK inhibitors have been recently published, showing an improved overall response rate. Short intracranial disease control is still a challenge. Several attempts have been made in order to improve it with combinations between local and systemic therapies. Immunotherapy approaches seem to retain promising activity in the treatment of melanoma brain metastasis as showed by the results of clinical trials investigating the combination of anti-CTL4 (Ipilimumab) and anti-PD1(Nivolumab). Studies about the combination or the sequential approach of target therapy and immunotherapy are ongoing, with immature results. Several clinical trials are ongoing trying to explore new approaches in order to overcome tumor resistance. At this moment the correct therapeutic choices for melanoma with intracranial involvement is still a challenge and new strategies are needed.
Collapse
Affiliation(s)
- Paolo Becco
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
| | - Susanna Gallo
- Ospedale Mauriziano Umberto I-Largo Turati 62, 10128 Torino, Italy
- Correspondence:
| | - Stefano Poletto
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
- Department of Oncology, University of Turin, 10124 Torino, Italy
| | - Mirko Pio Manlio Frascione
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
- Department of Oncology, University of Turin, 10124 Torino, Italy
| | - Luca Crotto
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
| | - Alessandro Zaccagna
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
| | - Luca Paruzzo
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
- Department of Oncology, University of Turin, 10124 Torino, Italy
| | - Daniela Caravelli
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
| | - Fabrizio Carnevale-Schianca
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
| | - Massimo Aglietta
- Istituto di Candiolo, FPO - IRCCS - Str. Prov.le 142, km 3,95, 10060 Candiolo, Italy; (P.B.); (S.P.); (M.P.M.F.); (L.C.); (A.Z.); (L.P.); (D.C.); (F.C.-S.); (M.A.)
- Department of Oncology, University of Turin, 10124 Torino, Italy
| |
Collapse
|
16
|
Carr MJ, Sun J, Eroglu Z, Zager JS. An evaluation of encorafenib for the treatment of melanoma. Expert Opin Pharmacother 2019; 21:155-161. [PMID: 31790307 DOI: 10.1080/14656566.2019.1694664] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: In the treatment of advanced BRAF-mutant melanoma, selective regulation of the MAPK pathway with BRAF and MEK inhibition has emerged as one of the mainstays of therapy.Areas covered: The authors present the current data on encorafenib as a compound, its pharmacokinetic and pharmacodynamics properties. This review includes current data on encorafenib therapy as a single agent as well as in combination with the MEK-inhibitor binimetinib and other systemic therapies.Expert opinion: BRAF inhibition with encorafenib exhibits substantial antitumor activity with less paradoxical MAPK pathway activation leading to treatment resistance. Combination therapy with MEK inhibitors improves response rate, progression-free survival, and overall survival in patients with BRAF-mutant metastatic melanoma compared to prior treatment regimens. Serious adverse events, including the development of cutaneous malignancies, are reported at lower rates with combination therapy, while less severe events such as pyrexia can be more common. Existing data is lacking for a recommendation of triplet therapy, although results from multiple ongoing trials are highly anticipated.
Collapse
Affiliation(s)
- Michael J Carr
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - James Sun
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
17
|
Lewin J, Sayers L, Kee D, Walpole I, Sanelli A, Te Marvelde L, Herschtal A, Spillane J, Gyorki D, Speakman D, Estall V, Donahoe S, Pohl M, Pope K, Chua M, Sandhu S, McArthur GA, McCormack CJ, Henderson M, Hicks RJ, Shackleton M. Surveillance imaging with FDG-PET/CT in the post-operative follow-up of stage 3 melanoma. Ann Oncol 2019; 29:1569-1574. [PMID: 29659679 DOI: 10.1093/annonc/mdy124] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background As early detection of recurrent melanoma maximizes treatment options, patients usually undergo post-operative imaging surveillance, increasingly with FDG-PET/CT (PET). To assess this, we evaluated stage 3 melanoma patients who underwent prospectively applied and sub-stage-specific schedules of PET surveillance. Patients and methods From 2009, patients with stage 3 melanoma routinely underwent PET +/- MRI brain scans via defined schedules based on sub-stage-specific relapse probabilities. Data were collected regarding patient characteristics and outcomes. Contingency analyses were carried out of imaging outcomes. Results One hundred and seventy patients (stage 3A: 34; 3B: 93; 3C: 43) underwent radiological surveillance. Relapses were identified in 65 (38%) patients, of which 45 (69%) were asymptomatic. False-positive imaging findings occurred in 7%, and 6% had treatable second (non-melanoma) malignancies. Positive predictive values (PPV) of individual scans were 56%-83%. Negative scans had predictive values of 89%-96% for true non-recurrence [negative predictive values (NPV)] until the next scan. A negative PET at 18 months had NPVs of 80%-84% for true non-recurrence at any time in the 47-month (median) follow-up period. Sensitivity and specificity of the overall approach of sub-stage-specific PET surveillance were 70% and 87%, respectively. Of relapsed patients, 33 (52%) underwent potentially curative resection and 10 (16%) remained disease-free after 24 months (median). Conclusions Application of sub-stage-specific PET in stage 3 melanoma enables asymptomatic detection of most recurrences, has high NPVs that may provide patient reassurance, and is associated with a high rate of detection of resectable and potentially curable disease at relapse.
Collapse
Affiliation(s)
- J Lewin
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Victoria, Australia; Princess Margaret Cancer Centre, Toronto, Canada
| | - L Sayers
- Cancer Treatment and Development Laboratory, Peter MacCallum Cancer Centre, Victoria, Australia
| | - D Kee
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Victoria, Australia
| | - I Walpole
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Victoria, Australia
| | - A Sanelli
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Victoria, Australia
| | - L Te Marvelde
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Victoria, Australia
| | - A Herschtal
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Victoria, Australia
| | - J Spillane
- Department of Surgery, Peter MacCallum Cancer Centre, Victoria, Australia
| | - D Gyorki
- Department of Surgery, Peter MacCallum Cancer Centre, Victoria, Australia; Department of Surgery, St Vincent's Hospital, Victoria, Australia
| | - D Speakman
- Department of Surgery, Peter MacCallum Cancer Centre, Victoria, Australia
| | - V Estall
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - S Donahoe
- Department of Surgery, Peter MacCallum Cancer Centre, Victoria, Australia
| | - M Pohl
- Department of Surgery, Peter MacCallum Cancer Centre, Victoria, Australia
| | - K Pope
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - M Chua
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - S Sandhu
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Victoria, Australia
| | - G A McArthur
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| | - C J McCormack
- Department of Dermatology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - M Henderson
- Department of Surgery, Peter MacCallum Cancer Centre, Victoria, Australia; Department of Surgery, St Vincent's Hospital, Victoria, Australia
| | - R J Hicks
- Department of Surgery, St Vincent's Hospital, Victoria, Australia; Department of Cancer Imaging, Peter MacCallum Cancer Centre, Victoria, Australia
| | - M Shackleton
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Victoria, Australia; Cancer Treatment and Development Laboratory, Peter MacCallum Cancer Centre, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Department of Pathology, The University of Melbourne, Victoria, Australia; Department of Oncology, Alfred Health, Victoria, Australia; Central Clinical School, Faculty of Medicine, Nursing and Allied Health, Monash University, Victoria, Australia.
| |
Collapse
|
18
|
Abstract
Targeted BRAF and MEK inhibition has become an appropriate first-line treatment of BRAF-mutant advanced cutaneous melanoma. The authors present an overview of the MAPK pathway as well as the other major pathways implicated in melanoma development. Melanoma brain metastases are a devastating complication of melanoma that can be traced to derangements in cell signaling pathways, and the current evidence for targeted therapy is reviewed. Finally, activating KIT mutations are rarely found to cause melanomas and may provide an actionable target for therapy. The authors review the current evidence for targeted KIT therapy and summarize the ongoing clinical trials.
Collapse
Affiliation(s)
- James Sun
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, 4th Floor, Tampa, FL 33612, USA
| | - Michael J Carr
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, 4th Floor, Tampa, FL 33612, USA
| | - Nikhil I Khushalani
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, 4th Floor, Tampa, FL 33612, USA.
| |
Collapse
|
19
|
Guida M, Bartolomeo N, De Risi I, Fucci L, Armenio A, Filannino R, Ruggieri E, Macina F, Traversa M, Nardone A, Figliuolo F, De Luca F, Mele F, Tommasi S, Strippoli S. The Management of Oligoprogression in the Landscape of New Therapies for Metastatic Melanoma. Cancers (Basel) 2019; 11:cancers11101559. [PMID: 31615127 PMCID: PMC6826412 DOI: 10.3390/cancers11101559] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/03/2019] [Accepted: 10/12/2019] [Indexed: 12/15/2022] Open
Abstract
Background: A limited degree of progression after a response to treatment is labelled as oligoprogression and is a hot topic of metastatic melanoma (MM) management. Rogue progressive metastases could benefit from local treatment, which could allow the continuation of ongoing systemic therapy, also known as treatment beyond progression (TBP). Methods: We retrospectively reviewed 214 selected MM patients who developed oligoprogression during treatment with v-Raf murine sarcoma viral oncogene homolog B (BRAF)/mitogen-activated-extracellular signal-regulated kinase (MEK) or programmed cell death protein 1 (PD-1) inhibitors and received a local treatment continuing TBP. We performed univariate and multivariable analyses to assess the association between therapy outcomes and a series of clinical and biological features. Results: We identified 27 (10%) oligoprogressed patients treated locally with surgery (14), radiosurgery (11), and electrochemotherapy (2). TBP included PD-1 inhibitors (13) and BRAF/MEK inhibitors (14). The median progression-free survival post oligoprogression (PFSPO) was 14 months (5-19 95% confidence interval (C.I.)). In the univariate analysis, a significantly longer PFSPO was associated with complete response (CR), Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 0, neutrophils/lymphocytes ratio (N/L) <2, and progression-free survival (PFS) at oligoprogression >11 months. Nevertheless, in the multivariable analysis, only CR and N/L <2 were found to be associated with longer PFSPO. Conclusions: In selected patients, local treatments contribute to controlling oligoprogression for a long time, allowing the continuation of systemic treatment and prolongation of overall survival (OS). Increasing biological and clinical knowledge is improving the accuracy in identifying patients to apply for local ablative therapies.
Collapse
Affiliation(s)
- Michele Guida
- Medical Oncology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Nicola Bartolomeo
- Department of Biomedical Sciences and Human Oncology, University of Bari, 70124 Bari, Italy.
| | - Ivana De Risi
- Medical Oncology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Livia Fucci
- Pathology Department National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Andrea Armenio
- Department of Plastic Surgery, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Ruggero Filannino
- Medical Oncology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Eustachio Ruggieri
- Department of Surgery, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Francesco Macina
- Radiology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Michele Traversa
- Radiology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Annalisa Nardone
- Radiology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Francesco Figliuolo
- Department of Plastic Surgery, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Federica De Luca
- Radiology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Fabio Mele
- Pathology Department National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| | - Stefania Tommasi
- Molecular Diagnostic and Pharmacogenetics laboratory, National Cancer Research Centre "Giovanni Paolo II", Bari 70124, Italy.
| | - Sabino Strippoli
- Medical Oncology Department, National Cancer Research Centre "Giovanni Paolo II", 70124 Bari, Italy.
| |
Collapse
|
20
|
Howard MD, Wee E, Wolfe R, McLean CA, Kelly JW, Pan Y. Anatomic location of primary melanoma: Survival differences and sun exposure. J Am Acad Dermatol 2019; 81:500-509. [DOI: 10.1016/j.jaad.2019.04.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/03/2019] [Accepted: 04/14/2019] [Indexed: 10/27/2022]
|
21
|
Vrielink OM, Kruijff S, van Leeuwen BL, Roodenburg JL. Application of CO 2 laser evaporation in locally advanced melanoma. Melanoma Manag 2019; 6:MMT14. [PMID: 31236206 PMCID: PMC6582456 DOI: 10.2217/mmt-2018-0008] [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: 11/02/2018] [Accepted: 02/18/2019] [Indexed: 11/21/2022] Open
Abstract
Aim This study aims to investigate the role of CO2 laser evaporation in the treatment of melanoma patients with satellite or in-transit metastases. Materials & methods Patients who underwent CO2 laser evaporation were retrospectively included between November 2002 and August 2018. The Sharplan 40C CO2 laser was used with a high pulse wave mode. Data concerning patient and tumor characteristics, CO2 laser evaporation and subsequent therapies were collected. Results A total of 26 patients were included. Median duration of local control was 5.5 months. The median number of lesions evaporated per treatment was three (1-16); patients received a median of three (1-19) treatments. Conclusion In a selected group of melanoma patients with satellite or in-transit metastases, CO2 laser evaporation should be considered as treatment for local control.
Collapse
Affiliation(s)
- Otis M Vrielink
- Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Schelto Kruijff
- Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Barbara L van Leeuwen
- Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Ln Roodenburg
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Perone JA, Farrow N, Tyler DS, Beasley GM. Contemporary Approaches to In-Transit Melanoma. J Oncol Pract 2019; 14:292-300. [PMID: 29746804 DOI: 10.1200/jop.18.00063] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In-transit melanoma represents a distinct disease pattern of heterogeneous superficial tumors. Many treatments have been developed specifically for this type of disease, including regional chemotherapy and a variety of directly injectable agents. Novel strategies include the intralesional delivery of oncolytic viruses and immunocytokines. The combination of intralesional or regional chemotherapy with systemic immune checkpoint inhibitors also is a promising approach. In the current review, we examine the general management of the workup of patients with in-transit disease, the range of available therapies, and recommendations for specific therapies for an individual patient.
Collapse
Affiliation(s)
- Jennifer A Perone
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Nellie Farrow
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Douglas S Tyler
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Georgia M Beasley
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| |
Collapse
|
23
|
Activation of TAp73 and inhibition of TrxR by Verteporfin for improved cancer therapy in TP53 mutant pancreatic tumors. Future Sci OA 2019; 5:FSO366. [PMID: 30820346 PMCID: PMC6391631 DOI: 10.4155/fsoa-2018-0082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
Aim: TAp73 is a tumor suppressor, which compensates for p53 loss and induces apoptosis in tumors in response to genotoxic stress or small-molecule treatments. Pancreatic ductal adenocarcinoma has a late onset of the disease, responds poorly to the existing therapies and has a very low survival rates. Result: Here, using drug-repurposing approach, we found that protoporphyrin IX (PpIX) and benzoporphyrin derivative (BPD) monoacid ring A activate TAp73 and induce apoptosis in pancreatic cancer cells. PpIX and BPD induce reactive oxygen species and inhibit thioredoxin reductase 1. Conclusion: Thus, PpIX and BPD target cancer cells’ vulnerabilities namely activate TAp73 tumor suppressor and inhibit oncogenic Trx1. Our findings may contribute to faster repurposing of PpIX and BPD to treat pancreatic tumors. Despite all efforts, pancreatic cancer remains one of the most aggressive tumors. Late diagnoses, often linked with the asymptomatic disease progression, make pancreatic cancer extremely difficult to treat. We have assessed drugs that are already clinically used in the photodynamic therapy of actinic keratosis and age-related macular degeneration and showed that the compounds induce apoptotic death of cancer cells. The mechanism is via activation of the p73 tumor suppressor and inhibition of the oncogenic thioredoxin reductase. Molecules with a complementary mechanism of action inducing cell death might be very promising candidates for improved cancer therapy in pancreatic cancer patients.
Two pathways leading to the induction of potent apoptosis by protoporphyrin IX and Verteporfin in mutant TP53 pancreatic cancer cells. One pathway is through inhibition of TAp73/MDM2 interactions and induction of TAp73-related apoptosis and the second pathway converges on inhibition of oncogenic TrxR and accumulation of reactive oxygen species.
Collapse
|
24
|
Prognostic significance of distant metastasis-free interval in patients with relapsed melanoma treated with BRAF with or without MEK inhibitors. Melanoma Res 2019; 29:428-434. [PMID: 30601377 DOI: 10.1097/cmr.0000000000000562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This retrospective cohort study assessed the prognostic significance of distant metastasis-free interval (DMFI) in patients with relapsed BRAF-mutant melanoma treated with BRAF with or without MEK inhibitors (BRAFi ± MEKi). Patients with a DMFI of up to 24 months were compared with those with DMFI of more than 24 months, with regard to their postrelapse progression-free survival (PR-PFS) and overall survival (PR-OS). In total, 109 patients were included in the study. Median DMFI was 25.3 (range: 3.4-188.2) months. Median PR-PFS in patients with DMFI of more than 24 months was 7.9 months [95% confidence interval (CI): 6.2-9.7] compared with 5.4 (95% CI: 4.2-6.7) months of those with shorter DMFI (P = 0.016). Median PR-OS was 15.6 months (95% CI: 13.6-17.6) in patients with DMFI of more than 24 months and 12.0 months (95% CI: 9.0-15.0) with DMFI of up to 24 months (P = 0.289). Multivariate Cox regression analysis showed that DMFI was independently and strongly associated with improved PR-PFS (adjusted hazard ratio = 3.21, 95% CI: 1.78-5.77, ≤ 24 vs. > 24 months) and longer PR-OS (adjusted hazard ratio: 2.09, 95% CI: 1.15-3.80, ≤ 24 vs. > 24 months). The present cohort study is one of the first to confirm the association of DMFI of more than 24 months with an indolent disease course, as shown by longer PR-PFS and PR-OS, in patients with relapsed stage IV melanoma treated by BRAF inhibitor/MEK inhibitor.
Collapse
|
25
|
Tawbi HA, Boutros C, Kok D, Robert C, McArthur G. New Era in the Management of Melanoma Brain Metastases. Am Soc Clin Oncol Educ Book 2018; 38:741-750. [PMID: 30231345 DOI: 10.1200/edbk_200819] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The remarkable advances in the systemic therapy of metastatic melanoma have now extended the 1-year overall survival rate from 25% to nearing 85%. Systemic treatment in the form of BRAF-targeted therapy and immunotherapy is slowly but surely proving its efficacy in the treatment of metatstatic brain metastases (MBM). Single-agent BRAF inhibitors provide an intracranial response rate of 25% to 40%, whereas the combination of BRAFi/MEKi leads to responses in up to 58%. However, the durability of responses induced by BRAFi/MEKi seems to be even shorter than in extracranial disease. On the other hand, single-agent ipilimumab provides comparable clinical benefit in MBMs as it does in extracranial metastases. Single-agent PD-1 anitbodies induce response rates of approximately 20%, and those responses appear durable. Similarly the combination of CTLA-4+ PD-1 antibodies induces durable responses at an impressive rate of 55% and is safe to administer. Although the local treatment approaches with radiation and surgery remain important and are critically needed in the management of MBM, systemic therapy offers a new dimension that can augment the impact of those therapies and come at a potentially lower cost of neurocognitive impairment. Considerations for combining those modalities are direly needed, in addition to considering novel systemic combinations that target mechanisms specific to MBM. In this report, we will discuss the underlying biology of melanoma brain metastases, the clinical outcomes from recent clinical trials of targeted and immunotherapy, and their impact on clinical practice in the context of existing local therapeutic modalities.
Collapse
Affiliation(s)
- Hussein A Tawbi
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Institut Gustave Roussy, Paris, France; Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Celine Boutros
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Institut Gustave Roussy, Paris, France; Peter MacCallum Cancer Centre, Melbourne, Australia
| | - David Kok
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Institut Gustave Roussy, Paris, France; Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Caroline Robert
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Institut Gustave Roussy, Paris, France; Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Grant McArthur
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Institut Gustave Roussy, Paris, France; Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
26
|
Jabbarzadeh Kaboli P, Ismail P, Ling KH. Molecular modeling, dynamics simulations, and binding efficiency of berberine derivatives: A new group of RAF inhibitors for cancer treatment. PLoS One 2018; 13:e0193941. [PMID: 29565994 PMCID: PMC5863970 DOI: 10.1371/journal.pone.0193941] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/21/2018] [Indexed: 12/19/2022] Open
Abstract
RAF kinases are a family of enzymes in the MAP kinase pathway that contribute to the development of different types of cancer. BRAF is the most important member of RAF kinases. BRAF mutations have been detected in 7% of all cancers and 66% of melanomas; as such, the FDA has approved a few BRAF inhibitor drugs to date. However, BRAF can activate CRAF leading to resistance to BRAF inhibitors. Berberine (BBR) is an alkaloid that is widely distributed in different plant species. Several studies have been carried out on the anti-cancer effects of BBR but direct targets of BBR are unknown. In this study, interactions of BBR derivatives against BRAF and CRAF kinases were modeled and predicted using an in silico-based approach. To analyze and identify the residues important in BRAF docking, we modeled interactions of ATP, the universal substrate of BRAF, and found that Lys483 and Asp594 are the most important residues involved in both ATP and BBR binding [(The average score = -11.5 kcal/mol (ATP); Range of scores = -7.78 to -9.55 kcal/mol (BBR)]. In addition to these polar residues, Trp530 and Phe583 are also applicable to the molecular docking of BRAF. We also observed that Asp593 was excluded from the enzyme cavity, while Phe594 was included inside the cavity, making the enzyme inactive. Finally, three alternatives for BBR were identified with dual RAF inhibition effects [The best scores against BRAF = -11.62 kcal/mol (BBR-7), -10.64 kcal/mol (BBR-9), and -11.01 kcal/mol (BBR-10); the best scores against CRAF = -9.68 kcal/mol (BBR-7), -9.60 kcal/mol (BBR-9), and -9.20 kcal/mol (BBR-10)]. Direct effects of BBR derivatives against BRAF and CRAF kinases had not yet been reported previously, and, thus, for the first time, we report three cycloprotoberberines as lead compounds against RAF kinases.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Patimah Ismail
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- * E-mail:
| | - King-Hwa Ling
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
27
|
Long-term survival with modern therapeutic agents against metastatic melanoma-vemurafenib and ipilimumab in a daily life setting. Med Oncol 2018; 35:24. [PMID: 29387968 DOI: 10.1007/s12032-018-1084-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Despite new therapeutic options, metastatic melanoma remains to be one of the most fatal tumors. With the development of BRAF inhibitors and immune checkpoint inhibitors, overall survival could be prolonged significantly for the first time. Clinical studies implied that even long-term survival is possible with both types of drugs, but predictive markers are so far missing. In this study, we analyzed survival data from patients that received the first-in-class substances vemurafenib and ipilimumab, respectively, during the time period from registration of the drugs until availability of combination treatments. We aimed to evaluate the possibility of long-term survival in a daily life setting and to characterize patients that benefit from these drugs in order to gain insight into predictive attributes. Eighty patients were evaluated who were treated with either vemurafenib (n = 40) or ipilimumab (n = 40), and overall survival was analyzed. Subgroup analysis was performed for patients who were still alive 24 months after induction of therapy (long-term survival). Median overall survival (OS) was 8.0 months for patients treated with vemurafenib and 10.0 months for patients treated with ipilimumab (log-rank P value = 0.689). Long-term survival was achieved in 32.5% of patients (42.3% vemurafenib, 57.7% ipilimumab). Negative predictors of long-term survival in the vemurafenib group were brain and liver metastases, as well as elevated LDH, S100ß and liver enzymes. For ipilimumab, an increase in lymphocytes and eosinophils during course of treatment correlated with long-term survival. Our real-life experience shows that long-term survival is possible with using both therapeutic agents, vemurafenib and ipilimumab. Pattern of metastases and laboratory values might be of interest in decision making for a specific therapeutic approach. Combination of drugs and observational studies in larger patient cohorts are necessary to further validate our findings.
Collapse
|
28
|
A Retrospective Analysis of the Efficacy of Pembrolizumab in Melanoma Patients With Brain Metastasis. J Immunother 2018; 40:108-113. [PMID: 28221189 DOI: 10.1097/cji.0000000000000159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A total of 50% of patients with melanoma will develop brain metastasis (BM). Pembrolizumab was approved for treatment of metastatic melanoma on the basis of significant systemic antitumor activity. Because of low enrollment of patients with BM in pembrolizumab trials, efficacy against melanoma BM remains unknown. We reviewed records of 89 consecutive patients with melanoma treated with pembrolizumab at our institution between May 1, 2014 and October 31, 2015 to determine the time to progression. Thirty-six (40%) patients had BM before pembrolizumab. Twenty-six (72%) patients with BM had received prior treatment for BM. With median follow-up of 17.2 months, 54 patients (61%) developed progressive disease on pembrolizumab. Intracranial progression occurred in 19 patients (21%), 3 of whom did not have BM before treatment. Median time to progression at any site was 6 months for those without BM (n=53), 5 months for those with treated BM (n=26), and 1.2 months for patients with untreated BM (n=10). Using a Cox regression model adjusted for baseline factors, there was a statistically significant (Wald χ P=0.003) reduction in the hazard of progression for patients without BM [hazard ratio, 0.19; 90% confidence interval, 0.08-0.42) and patients with treated BM (hazard ratio, 0.27; 90% confidence interval, 0.12-0.64) compared with those with untreated BM. In conclusion, melanoma patients with pretreated BM can have durable systemic responses to pembrolizumab. Large, prospective studies are needed to evaluate the intracranial antitumor activity of pembrolizumab in melanoma patients with untreated BM.
Collapse
|
29
|
Hassel JC, Buder‐Bakhaya K, Bender C, Zimmer L, Weide B, Loquai C, Ugurel S, Slynko A, Gutzmer R, the German Dermatooncology Group (DeCOG/ADO). Progression patterns under BRAF inhibitor treatment and treatment beyond progression in patients with metastatic melanoma. Cancer Med 2018; 7:95-104. [PMID: 29266761 PMCID: PMC5773979 DOI: 10.1002/cam4.1267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/22/2017] [Accepted: 10/30/2017] [Indexed: 01/07/2023] Open
Abstract
Despite markedly improved treatment options for metastatic melanoma, resistance to targeted therapies such as BRAF inhibitors (BRAFi) or BRAFi plus MEK inhibitors (MEKi) remains a major problem. Our aim was to characterize progression on BRAFi therapy and outcome of subsequent treatment. One hundred and eighty patients with BRAF-mutant metastatic melanoma who had progressed on treatment with single-agent BRAFi from February 2010 to April 2015 were included in a retrospective data analysis focused on patterns of progression, treatment beyond progression (TBP) and subsequent treatments after BRAFi therapy. Analysis revealed that 51.1% of patients progressed with both new and existing metastases opposed to progression of only preexisting (28.3%) or only new (20.6%) metastases. Exclusive extracranial progression occurred in 50.6% of patients compared to both extra- and intracranial (29.4%) or sole cerebral progression (20%). Multivariable analyses demonstrated that single site progression and primary response to BRAFi were associated with improved progression-free survival. Progression with exclusively new or only existing metastases and a baseline Eastern Cooperative Oncology Group (ECOG) of 0 were associated with prolonged overall survival (OS). TBP had no significant impact on OS. Other subsequent treatments showed low efficacy with the exception of anti-PD-1 antibodies. In conclusion we identified specific patterns of progression which significantly correlate with further prognosis after progression on BRAFi treatment. In contrast to previously published data, we could not demonstrate a significant survival benefit for BRAFi TBP. Subsequent therapies had strikingly low efficacy except for PD-1 inhibitors.
Collapse
Affiliation(s)
- Jessica C. Hassel
- Department of Dermatology and National Center for Tumor DiseasesUniversity Hospital HeidelbergHeidelbergGermany
| | - Kristina Buder‐Bakhaya
- Department of Dermatology and National Center for Tumor DiseasesUniversity Hospital HeidelbergHeidelbergGermany
| | - Carolin Bender
- Department of Dermatology and National Center for Tumor DiseasesUniversity Hospital HeidelbergHeidelbergGermany
| | - Lisa Zimmer
- Department of DermatologyUniversity Hospital EssenUniversity Duisburg‐EssenEssenGermany
| | - Benjamin Weide
- Department of DermatologyCenter for DermatooncologyUniversity Medical Center TübingenTübingenGermany
| | - Carmen Loquai
- Department of DermatologyUniversity Medical CenterJohannes Gutenberg‐UniversityMainzGermany
| | - Selma Ugurel
- Department of DermatologyUniversity Hospital EssenUniversity Duisburg‐EssenEssenGermany
| | - Alla Slynko
- Department of Mathematics, Natural and Economic SciencesUniversity of Applied SciencesUlmGermany
| | - Ralf Gutzmer
- Department of Dermatology and AllergySkin Cancer Center HannoverHannover Medical SchoolHannoverGermany
| | | |
Collapse
|
30
|
Peng L, Wang Y, Hong Y, Ye X, Shi P, Zhang J, Zhao Q. Incidence and relative risk of cutaneous squamous cell carcinoma with single-agent BRAF inhibitor and dual BRAF/MEK inhibitors in cancer patients: a meta-analysis. Oncotarget 2017; 8:83280-83291. [PMID: 29137342 PMCID: PMC5669968 DOI: 10.18632/oncotarget.21059] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/28/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND BRAF inhibitor and dual BRAF/MEK inhibitors have been approved for the treatment of BRAF-mutated melanoma. Cutaneous squamous cell carcinoma (cuSCC) is an adverse event associated with these drugs. The contribution of BRAF inhibitor and dual BRAF/MEK inhibitors to cuSCC are still unknown. We performed this meta-analysis to determine the overall incidence and relative risk of cuSCC in cancer patients treated with these drugs. RESULTS A total of 7,442 patients from 24 primary studies were included. The incidences of all-grade and high-grade cuSCC in cancer patients treated with BRAF inhibitor were 12.5% (95% CI: 10.8-14.6%) and 11.6% (95% CI: 9.8-13.8%), and dual BRAF/MEK inhibitors were 3.0% (95% CI: 2.0-4.5%) and 2.8% (95% CI: 1.9-4.0%), respectively. On subgroup analysis and meta-regression, the incidence of cuSCC did not vary with tumor type, study design and specific drug used. The use of single agent BRAF inhibitor significantly increased the risk of developing cuSCC comparing with dual BRAF/MEK inhibitors for all-grade (RR 4.72, 95% CI: 2.42-9.20) and high-grade (RR 4.92, 95% CI: 2.64-9.16) in cancer patients. MATERIALS AND METHODS The databases of PubMed, Embase and abstracts published in ASCO proceedings were searched for relevant studies from January 2000 to June 2017. Summary incidences, relative risks (RRs) and 95% confidence intervals (CIs) were calculated by using either random effects or fixed effect models according to the heterogeneity of included studies. CONCLUSIONS BRAF inhibitor significantly increases the risk of developing cuSCC compared with dual BRAF/MEK inhibitors in cancer patients. Clinicians should be aware of the risks of cuSCC with the administration of these drugs in cancer patients.
Collapse
Affiliation(s)
- Ling Peng
- Department of Thoracic Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yina Wang
- Department of Thoracic Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yun Hong
- Department of Pharmacy, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xianghua Ye
- Department of Radiotherapy, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Peng Shi
- Department of Medical Statistics, Children’s Hospital of Fudan University, Shanghai, China
- Center for Evidence-based Medicine, Fudan University, Shanghai, China
| | - Junyan Zhang
- Bothwin Clinical Study Consultant, Bellevue, WA, USA
| | - Qiong Zhao
- Department of Thoracic Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
31
|
Arozarena I, Wellbrock C. Overcoming resistance to BRAF inhibitors. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:387. [PMID: 29114545 PMCID: PMC5653517 DOI: 10.21037/atm.2017.06.09] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
The discovery of activating mutations in the serine/threonine (S/T) kinase BRAF followed by a wave of follow-up research manifested that the MAPK-pathway plays a critical role in melanoma initiation and progression. BRAF and MEK inhibitors produce an unparalleled response rate in melanoma, but it is now clear that most responses are transient, and while some patients show long lasting responses the majority progress within 1 year. In accordance with the key role played by the MAPK-pathway in BRAF mutant melanomas, disease progression is mostly due to the appearance of drug-resistance mechanisms leading to restoration of MAPK-pathway activity. In the present article we will review the development, application and clinical effects of BRAF and MEK inhibitors both, as single agent and in combination in the context of targeted therapy in melanoma. We will then describe the most prominent mechanisms of resistance found in patients progressed on these targeted therapies. Finally we will discuss strategies for further optimizing the use of MAPK inhibitors and will describe the potential of alternative combination therapies to either delay the onset of resistance to MAPK inhibitors or directly target specific mechanisms of resistance to BRAF/MEK inhibitors.
Collapse
Affiliation(s)
- Imanol Arozarena
- Navarrabiomed-Fundación Miguel Servet-Idisna, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
32
|
Hu-Lieskovan S, Ribas A. New Combination Strategies Using Programmed Cell Death 1/Programmed Cell Death Ligand 1 Checkpoint Inhibitors as a Backbone. Cancer J 2017; 23:10-22. [PMID: 28114250 PMCID: PMC5844278 DOI: 10.1097/ppo.0000000000000246] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery of immune checkpoints and subsequent clinical development of checkpoint inhibitors have revolutionized the field of oncology. The durability of the antitumor immune responses has raised the hope for long-term patient survival and potential cure; however, currently, only a minority of patients respond. Combination strategies to help increase antigen release and T-cell priming, promote T-cell activation and homing, and improve the tumor immune microenvironment, all guided by predictive biomarkers, can help overcome the tumor immune-evasive mechanisms and maximize efficacy to ultimately benefit the majority of patients. Great challenges remain because of the complex underlying biology, unpredictable toxicity, and accurate assessment of response. Carefully designed clinical trials guided by translational studies of paired biopsies will be key to develop reliable predictive biomarkers to choose which patients would most likely benefit from each strategy.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- From the Division of Hematology-Oncology, Department of Medicine, Jonsson Comprehensive Cancer Center at the University of California Los Angeles, Los Angeles, CA
| | | |
Collapse
|
33
|
Aya-Bonilla CA, Marsavela G, Freeman JB, Lomma C, Frank MH, Khattak MA, Meniawy TM, Millward M, Warkiani ME, Gray ES, Ziman M. Isolation and detection of circulating tumour cells from metastatic melanoma patients using a slanted spiral microfluidic device. Oncotarget 2017; 8:67355-67368. [PMID: 28978038 PMCID: PMC5620178 DOI: 10.18632/oncotarget.18641] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
Circulating Tumour Cells (CTCs) are promising cancer biomarkers. Several methods have been developed to isolate CTCs from blood samples. However, the isolation of melanoma CTCs is very challenging as a result of their extraordinary heterogeneity, which has hindered their biological and clinical study. Thus, methods that isolate CTCs based on their physical properties, rather than surface marker expression, such as microfluidic devices, are greatly needed in melanoma. Here, we assessed the ability of the slanted spiral microfluidic device to isolate melanoma CTCs via label-free enrichment. We demonstrated that this device yields recovery rates of spiked melanoma cells of over 80% and 55%, after one or two rounds of enrichment, respectively. Concurrently, a two to three log reduction of white blood cells was achieved with one or two rounds of enrichment, respectively. We characterised the isolated CTCs using multimarker flow cytometry, immunocytochemistry and gene expression. The results demonstrated that CTCs from metastatic melanoma patients were highly heterogeneous and commonly expressed stem-like markers such as PAX3 and ABCB5. The implementation of the slanted microfluidic device for melanoma CTC isolation enables further understanding of the biology of melanoma metastasis for biomarker development and to inform future treatment approaches.
Collapse
Affiliation(s)
- Carlos A Aya-Bonilla
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Gabriela Marsavela
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - James B Freeman
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Chris Lomma
- Department of Health, Perth, Western Australia, Australia
| | - Markus H Frank
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia.,Transplantation Research Program, Boston Children's Hospital and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Muhammad A Khattak
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia.,School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Tarek M Meniawy
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Michael Millward
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Majid E Warkiani
- School of Mechanical and Manufacturing Engineering, Australian Center for NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Mel Ziman
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia.,School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
34
|
Blank CU, Larkin J, Arance AM, Hauschild A, Queirolo P, Del Vecchio M, Ascierto PA, Krajsova I, Schachter J, Neyns B, Garbe C, Chiarion Sileni V, Mandalà M, Gogas H, Espinosa E, Hospers GAP, Miller WH, Robson S, Makrutzki M, Antic V, Brown MP. Open-label, multicentre safety study of vemurafenib in 3219 patients with BRAF V600 mutation-positive metastatic melanoma: 2-year follow-up data and long-term responders' analysis. Eur J Cancer 2017; 79:176-184. [PMID: 28501764 DOI: 10.1016/j.ejca.2017.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 03/16/2017] [Accepted: 04/07/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND The orally available BRAF kinase inhibitor vemurafenib is an effective and tolerable treatment option for patients with metastatic melanoma harbouring BRAFV600 mutations. We assessed the safety of vemurafenib in a large population of patients with few alternative treatment options; we report updated 2-year safety. METHODS This was an open-label, multicentre study of vemurafenib (960 mg bid) in patients with previously treated or untreated BRAF mutation-positive metastatic melanoma (cobas® 4800 BRAF V600 Mutation Test). The primary end-point was safety; efficacy end-points were secondary. An exploratory analysis was performed to assess safety outcomes in patients with long duration of response (DOR) (≥12 or ≥24 months). RESULTS After a median follow-up of 32.2 months (95% CI, 31.1-33.2 months), 3079/3219 patients (96%) had discontinued treatment. Adverse events (AEs) were largely consistent with previous reports; the most common all-grade treatment-related AEs were arthralgia (37%), alopecia (25%) and hyperkeratosis (23%); the most common grade 3/4 treatment-related AEs were squamous cell carcinoma of the skin (8%) and keratoacanthoma (8%). In the exploratory analysis, patients with DOR ≥12 months (n = 287) or ≥24 months (n = 133) were more likely to experience grade 3/4 AEs than the overall population. No new specific safety signals were observed with longer vemurafenib exposure. CONCLUSIONS After 2 years' follow-up, safety was maintained in this large group of patients with BRAFV600 mutation-positive metastatic melanoma who are more representative of routine clinical practice than typical clinical trial populations. These data suggest that long-term vemurafenib treatment is effective and tolerable without the development of new safety signals.
Collapse
Affiliation(s)
- Christian U Blank
- The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066CX, The Netherlands.
| | - James Larkin
- The Royal Marsden NHS Foundation Trust, London, UK.
| | - Ana M Arance
- Department of Medical Oncology, Hospital Clínic Barcelona, Barcelona, Spain.
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany.
| | | | - Michele Del Vecchio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | | | - Ivana Krajsova
- General University Hospital, Dermatooncology U, Prague, Czech Republic.
| | - Jacob Schachter
- Chaim Sheba Medical Centre, Oncology Institute, Ramat-Gan, Israel.
| | - Bart Neyns
- Afdelingshoofd, Medische Oncologie, Brussels, Belgium.
| | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany.
| | | | | | | | | | - Geke A P Hospers
- University Medical Centre Groningen, Groningen, The Netherlands.
| | - Wilson H Miller
- McGill University, Segal Cancer Centre, Montreal, Quebec, Canada.
| | | | | | | | - Michael P Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, Australia; Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia; Discipline of Medicine, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
35
|
Integrin β1 activation induces an anti-melanoma host response. PLoS One 2017; 12:e0175300. [PMID: 28448494 PMCID: PMC5407755 DOI: 10.1371/journal.pone.0175300] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/23/2017] [Indexed: 11/29/2022] Open
Abstract
TGF-β is a cytokine thought to function as a tumor promoter in advanced malignancies. In this setting, TGF-β increases cancer cell proliferation, survival, and migration, and orchestrates complex, pro-tumorigenic changes in the tumor microenvironment. Here, we find that in melanoma, integrin β1-mediated TGF-β activation may also produce tumor suppression via an altered host response. In the A375 human melanoma cell nu/nu xenograft model, we demonstrate that cell surface integrin β1-activation increases TGF-β activity, resulting in stromal activation, neo-angiogenesis and, unexpectedly for this nude mouse model, increase in the number of intra-tumoral CD8+ T lymphocytes within the tumor microenvironment. This is associated with attenuation of tumor growth and long-term survival benefit. Correspondingly, in human melanomas, TGF-β1 correlates with integrin β1/TGF-β1 activation and the expression of markers for vasculature and stromal activation. Surprisingly, this integrin β1/TGF-β1 transcriptional footprint also correlates with the expression of markers for tumor-infiltrating lymphocytes, multiple immune checkpoints and regulatory pathways, and, importantly, better long-term survival of patients. These correlations are unique to melanoma, in that we do not observe similar associations between β1 integrin/TGF-β1 activation and better long-term survival in other human tumor types. These results suggest that activation of TGF-β1 in melanoma may be associated with the generation of an anti-tumor host response that warrants further study.
Collapse
|
36
|
Escudier B, Motzer RJ, Sharma P, Wagstaff J, Plimack ER, Hammers HJ, Donskov F, Gurney H, Sosman JA, Zalewski PG, Harmenberg U, McDermott DF, Choueiri TK, Richardet M, Tomita Y, Ravaud A, Doan J, Zhao H, Hardy H, George S. Treatment Beyond Progression in Patients with Advanced Renal Cell Carcinoma Treated with Nivolumab in CheckMate 025. Eur Urol 2017; 72:368-376. [PMID: 28410865 DOI: 10.1016/j.eururo.2017.03.037] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/24/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Response patterns to nivolumab differ from those seen with other approved targeted therapies. OBJECTIVE To investigate the efficacy of nivolumab in previously treated patients with advanced renal cell carcinoma who were treated beyond (Response Evaluation Criteria In Solid Tumors) RECIST progression. DESIGN, SETTING, AND PARTICIPANTS This was a subgroup analysis of patients treated with nivolumab in the phase 3 CheckMate 025 study. Patients continuing to tolerate therapy and exhibiting investigator-assessed clinical benefit were eligible to be treated beyond RECIST progression (TBP) and received therapy for ≥4 wk after first progression; patients not treated beyond RECIST progression (NTBP) received 0 wk to <4 wk of therapy after progression. INTERVENTIONS Nivolumab 3mg/kg intravenously every 2 wk. RESULTS AND LIMITATIONS Of 406 nivolumab-treated patients, 316 (78%) progressed by RECIST criteria. Of those who progressed, 48% were TBP, 52% were NTBP. Before being TBP, objective response rate (95% confidence interval) was 20% (14-28) and 14% (9-21) in patients TBP and NTBP, respectively. Differences in clinical characteristics assessed at first progression between patients TBP versus NTBP included better Karnofsky performance status, less deterioration in Karnofsky performance status, shorter time to response, lower incidence of new bone lesions, and improved quality of life. Postprogression, 13% of all patients TBP (20/153) had ≥30% tumor burden reduction including patients with preprogression and postprogression tumor measurements (n=142) and complete/partial response (28%, 8/29), stable disease (6%, 3/47), and progressive disease (14%, 9/66) as their best response before being TBP. Incidence of treatment-related adverse events in patients TBP was lower after (59%) versus before (71%) progression. Limitations included potential bias from the nonrandomized nature of the analysis. CONCLUSIONS A subset of patients with advanced renal cell carcinoma and RECIST progression experienced tumor reduction postprogression with nivolumab, and had an acceptable safety profile. Clinical judgment remains essential when switching therapy. ClinicalTrials.gov Identifier: NCT01668784. PATIENT SUMMARY A subset of patients with advanced renal cell carcinoma and disease progression may continue to benefit from nivolumab treatment beyond progression as evidenced by tumor reduction postprogression and an acceptable safety profile.
Collapse
Affiliation(s)
- Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Villejuif, France.
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, Department of Immunology, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - John Wagstaff
- Department of Medical Oncology, South West Wales Cancer Institute and Swansea University College of Medicine, Swansea, UK
| | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hans J Hammers
- Department of Medical Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Howard Gurney
- Department of Medical Oncology, Westmead Hospital and Macquarie University, Sydney, Australia
| | - Jeffrey A Sosman
- Department of Hematology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pawel G Zalewski
- Department of Oncology, Lakeridge Health RS McLaughlin Durham Regional Cancer Centre, Oshawa, ON, Canada
| | - Ulrika Harmenberg
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - David F McDermott
- Biologic Therapy and Cutaneous Oncology Programs, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Toni K Choueiri
- Kidney Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA, USA
| | - Martin Richardet
- Department of Medical Oncology, Instituto Oncologico de Cordoba, Cordoba, Argentina
| | - Yoshihiko Tomita
- Department of Urology, Department of Molecular Oncology, Niigata University, Niigata, Japan
| | - Alain Ravaud
- Department of Medical Oncology and Radiotherapy, Bordeaux University Hospital, Hôpital Saint André, Bordeaux, France
| | | | | | | | - Saby George
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
37
|
McQuade JL, Posada LP, Lecagoonporn S, Cain S, Bassett RL, Patel SP, Hwu WJ, Hwu P, Davies MA, Bedikian AY, Amaria RN. A phase I study of TPI 287 in combination with temozolomide for patients with metastatic melanoma. Melanoma Res 2016; 26:604-608. [PMID: 27540836 PMCID: PMC5336360 DOI: 10.1097/cmr.0000000000000296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
TPI 287 is a synthetic taxane derivative with structural modifications allowing for central nervous system penetration and potential circumvention of multidrug resistance efflux pump mechanisms. The aim of this phase I study was to determine the maximum tolerated dose of the combination of TPI 287 and temozolomide in metastatic melanoma. Patients with stage IV unresectable or recurrent stage III melanoma were eligible. Stable untreated or treated brain metastases were allowed. Patients with previous taxane exposure were excluded. TPI 287 was administered intravenously on days 1, 8, and 15 and temozolomide was taken orally daily on days 1-5 of a 28-day cycle. Responses were assessed every two cycles according to WHO criteria. A total of 21 patients were enrolled. The maximum tolerated dose of the combination at this schedule was determined to be 125 mg/m intravenous of TPI 287 and 110 mg/m of oral temozolomide. The dose-limiting toxicity was neuropathy and six patients experienced grade III neuropathy. All patients were evaluable for tumor response. There were no complete responses; there were two partial responses and seven patients had stable disease (overall response rate 9.5% and disease control rate 42.9%). Three patients had stable disease in the brain despite progressive extracranial disease. The combination of TPI 287 and temozolomide is well tolerated in patients with metastatic melanoma, with the exception of neuropathy. The central nervous system penetration of both agents makes this a rational combination for further testing in primary and metastatic brain lesions.
Collapse
Affiliation(s)
- Jennifer L McQuade
- Departments of aMelanoma Medical Oncology bBiostatistics cSystems Biology, UT MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Dagogo-Jack I, Gill CM, Cahill DP, Santagata S, Brastianos PK. Treatment of brain metastases in the modern genomic era. Pharmacol Ther 2016; 170:64-72. [PMID: 27773784 DOI: 10.1016/j.pharmthera.2016.10.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Development of brain metastasis (BM) portends a dismal prognosis for patients with cancer. Melanomas and carcinomas of the lung, breast, and kidney are the most common malignancies to metastasize to the brain. Recent advances in molecular genetics have enabled the identification of actionable, clinically relevant genetic alterations within primary tumors and their corresponding metastases. Adoption of genotype-guided treatment strategies for the management of systemic malignancy has resulted in dramatic and durable responses. Unfortunately, despite these therapeutic advances, central nervous system (CNS) relapses are not uncommon. Although these relapses have historically been attributed to limited blood brain barrier penetration of anti-neoplastic agents, recent work has demonstrated genetic heterogeneity such that metastatic sites, including BM, harbor relevant genetic alterations that are not present in primary tumor biopsies. This improved insight into molecular mechanisms underlying site specific recurrences can inform strategies for targeting these oncogenic drivers. Thus, development of rational, genomically guided CNS-penetrant therapies is crucial for ongoing therapeutic success.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Corey M Gill
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Priscilla K Brastianos
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
39
|
Deniger DC, Kwong MLM, Pasetto A, Dudley ME, Wunderlich JR, Langhan MM, Lee CCR, Rosenberg SA. A Pilot Trial of the Combination of Vemurafenib with Adoptive Cell Therapy in Patients with Metastatic Melanoma. Clin Cancer Res 2016; 23:351-362. [PMID: 28093487 DOI: 10.1158/1078-0432.ccr-16-0906] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/14/2016] [Accepted: 09/23/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE This pilot feasibility clinical trial evaluated the coadministration of vemurafenib, a small-molecule antagonist of BRAFV600 mutations, and tumor-infiltrating lymphocytes (TIL) for the treatment of metastatic melanoma. EXPERIMENTAL DESIGN A metastatic tumor was resected for growth of TILs, and patients were treated with vemurafenib for 2 weeks, followed by resection of a second lesion. Patients then received a nonmyeloablative preconditioning regimen, infusion of autologous TILs, and high-dose interleukin-2 administration. Vemurafenib was restarted at the time of TIL infusion and was continued for 2 years or until disease progression. Clinical responses were evaluated by Response Evaluation Criteria in Solid Tumors (RECIST) 1.0. Metastases resected prior to and after 2 weeks of vemurafenib were compared using TCRB deep sequencing, immunohistochemistry, proliferation, and recognition of autologous tumor. RESULTS The treatment was well tolerated and had a safety profile similar to that of TIL or vemurafenib alone. Seven of 11 patients (64%) experienced an objective clinical response, and 2 patients (18%) had a complete response for 3 years (one response is ongoing at 46 months). Proliferation and viability of infusion bag TILs and peripheral blood T cells were inhibited in vitro by research-grade vemurafenib (PLX4032) when approaching the maximum serum concentration of vemurafenib. TCRB repertoire (clonotypes numbers, clonality, and frequency) did not significantly change between pre- and post-vemurafenib lesions. Recognition of autologous tumor by T cells was similar between TILs grown from pre- and post-vemurafenib metastases. CONCLUSIONS Coadministration of vemurafenib and TILs was safe and feasible and generated objective clinical responses in this small pilot clinical trial. Clin Cancer Res; 23(2); 351-62. ©2016 AACRSee related commentary by Cogdill et al., p. 327.
Collapse
Affiliation(s)
- Drew C Deniger
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mei Li M Kwong
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Anna Pasetto
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark E Dudley
- Cell and Gene Therapy, Novartis, Cambridge, Massachusetts
| | - John R Wunderlich
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michelle M Langhan
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Chyi-Chia Richard Lee
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Steven A Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
40
|
Fattore L, Mancini R, Acunzo M, Romano G, Laganà A, Pisanu ME, Malpicci D, Madonna G, Mallardo D, Capone M, Fulciniti F, Mazzucchelli L, Botti G, Croce CM, Ascierto PA, Ciliberto G. miR-579-3p controls melanoma progression and resistance to target therapy. Proc Natl Acad Sci U S A 2016; 113:E5005-E5013. [PMID: 27503895 PMCID: PMC5003278 DOI: 10.1073/pnas.1607753113] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Therapy of melanoma patients harboring activating mutations in the BRAF (V-raf murine sarcoma viral oncogene homolog B1) oncogene with a combination of BRAF and MEK inhibitors is plagued by the development of drug resistance. Mutational events, as well as adaptive mechanisms, contribute to the development of drug resistance. In this context we uncover here the role of a miRNA, miR-579-3p. We first show that low expression of miR-579-3p is a negative prognostic factor correlating with poor survival. Expression levels of miR-579-3p decrease from nevi to stage III/IV melanoma samples and even further in cell lines resistant to BRAF/MEK inhibitors. Mechanistically, we demonstrate that miR-579-3p acts as an oncosuppressor by targeting the 3'UTR of two oncoproteins: BRAF and an E3 ubiquitin protein ligase, MDM2. Moreover miR-579-3p ectopic expression impairs the establishment of drug resistance in human melanoma cells. Finally, miR-579-3p is strongly down-regulated in matched tumor samples from patients before and after the development of resistance to targeted therapies.
Collapse
Affiliation(s)
- Luigi Fattore
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples 80131, Italy
| | - Rita Mancini
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome 00161, Italy
| | - Mario Acunzo
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Giulia Romano
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Alessandro Laganà
- Department of Genetics, Genomic Sciences Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Maria Elena Pisanu
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Catanzaro "Magna Graecia", Catanzaro 88100, Italy
| | - Debora Malpicci
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Catanzaro "Magna Graecia", Catanzaro 88100, Italy
| | - Gabriele Madonna
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples 80131, Italy
| | - Domenico Mallardo
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples 80131, Italy
| | - Marilena Capone
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples 80131, Italy
| | - Franco Fulciniti
- Istituto Cantonale di Patologia, Servizio di Citologia Clinica, 6600 Locarno, Switzerland
| | - Luca Mazzucchelli
- Istituto Cantonale di Patologia, Servizio di Citologia Clinica, 6600 Locarno, Switzerland
| | - Gerardo Botti
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples 80131, Italy
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
| | - Paolo Antonio Ascierto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples 80131, Italy
| | - Gennaro Ciliberto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples 80131, Italy;
| |
Collapse
|
41
|
Ji C, Zhang Z, Chen L, Zhou K, Li D, Wang P, Huang S, Gong T, Cheng B. Endoplasmic reticulum stress-induced autophagy determines the susceptibility of melanoma cells to dabrafenib. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2491-8. [PMID: 27536070 PMCID: PMC4977101 DOI: 10.2147/dddt.s112740] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Melanoma is one of the deadliest skin cancers and accounts for most skin-related deaths due to strong resistance to chemotherapy drugs. In the present study, we investigated the mechanisms of dabrafenib-induced drug resistance in human melanoma cell lines A375 and MEL624. Our studies support that both endoplasmic reticulum (ER) stress and autophagy were induced in the melanoma cells after the treatment with dabrafenib. In addition, ER stress-induced autophagy protects melanoma cells from the toxicity of dabrafenib. Moreover, inhibition of both ER stress and autophagy promote the sensitivity of melanoma cells to dabrafenib. Taken together, the data suggest that ER stress-induced autophagy determines the sensitivity of melanoma cells to dabrafenib. These results provide us with promising evidence that the inhibition of autophagy and ER stress could serve a therapeutic effect for the conventional dabrafenib chemotherapy.
Collapse
Affiliation(s)
- Chao Ji
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Ziping Zhang
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Lihong Chen
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Kunli Zhou
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Dongjun Li
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Ping Wang
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Shuying Huang
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Ting Gong
- Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Bo Cheng
- Department of Dermatology, the 1st Affiliated Hospital of Fujian Medical University; Fujian Institute of Dermatology and Venereology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
42
|
Dolinsek T, Prosen L, Cemazar M, Potocnik T, Sersa G. Electrochemotherapy with bleomycin is effective in BRAF mutated melanoma cells and interacts with BRAF inhibitors. Radiol Oncol 2016; 50:274-9. [PMID: 27679543 PMCID: PMC5024665 DOI: 10.1515/raon-2016-0042] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The aim of the study was to explore the effectiveness of electrochemotherapy (ECT) during the treatment of melanoma patients with BRAF inhibitors. Its effectiveness was tested on BRAF mutated and non-mutated melanoma cells in vitro and in combination with BRAF inhibitors. MATERIALS AND METHODS ECT with bleomycin was performed on two human melanoma cell lines, with (SK-MEL-28) or without (CHL-1) BRAF V600E mutation. Cell survival was determined using clonogenic assay to determine the effectiveness of ECT in melanoma cells of different mutation status. Furthermore, the effectiveness of ECT in concomitant treatment with BRAF inhibitor vemurafenib was also determined in BRAF mutated cells SK-MEL-28 with clonogenic assay. RESULTS The survival of BRAF V600E mutated melanoma cells was even lower than non-mutated cells, indicating that ECT is effective regardless of the mutational status of melanoma cells. Furthermore, the synergistic interaction between vemurafenib and ECT with bleomycin was demonstrated in the BRAF V600E mutated melanoma cells. CONCLUSIONS The effectiveness of ECT in BRAF mutated melanoma cells as well as potentiation of its effectiveness during the treatment with vemurafenib in vitro implies on clinical applicability of ECT in melanoma patients with BRAF mutation and/or during the treatment with BRAF inhibitors.
Collapse
Affiliation(s)
| | - Lara Prosen
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Maja Cemazar
- Institute of Oncology Ljubljana, Ljubljana, Slovenia; Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | | | - Gregor Sersa
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
43
|
Combined vemurafenib and fotemustine in patients with BRAF V600 melanoma progressing on vemurafenib. Oncotarget 2016; 9:12408-12417. [PMID: 29552321 PMCID: PMC5844757 DOI: 10.18632/oncotarget.10589] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/30/2016] [Indexed: 01/08/2023] Open
Abstract
Background BRAF inhibitor vemurafenib achieves high response rate and an improvement in survival in patients with BRAF-mutated metastatic melanoma. However, median progression-free survival is only 6.9 months in the phase 3 study. Retrospective analyses suggest that treatment with BRAF inhibitors beyond initial progression might be associated with improved overall survival. We aimed to prospectively investigate the activity of prolonged treatment with vemurafenib and the addition of fotemustine in patients with systemic progression on prior single-agent BRAF inhibitor. Patients and Methods In this two-centres, single-arm Phase 2 trial, we enrolled patients with systemic progressive disease during single-agent vemurafenib treatment. Participants received vemurafenib 960 mg twice daily or dose administered at time of disease progression with vemurafenib previous treatment and fotemustine 100 mg/m2 intravenously every three weeks. The primary endpoint was PFS. Results Thirty-one patients were enrolled in the study; 16 patients had brain metastases at baseline. Median PFS was 3.9 months and 19 patients (61.3%) achieved disease control (1 CR, 4 PR, 14 SD). For patients achieving disease control, median duration of treatment was 6 months. Median OS was 5.8 months from enrolment and 15.4 months from start of previous vemurafenib. Five patients (16.1%) had a G3-4 AE, the most common being thrombocytopenia, which occurred in 3 patients. This trial is registered with ClinicalTrials.gov number NCT01983124. Conclusion The combination of vemurafenib plus fotemustine has clinical activity and an acceptable safety profile in BRAF-refractory patients.
Collapse
|
44
|
Dagogo-Jack I, Carter SL, Brastianos PK. Brain Metastasis: Clinical Implications of Branched Evolution. Trends Cancer 2016; 2:332-337. [DOI: 10.1016/j.trecan.2016.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022]
|
45
|
Kim A, Cohen MS. The discovery of vemurafenib for the treatment of BRAF-mutated metastatic melanoma. Expert Opin Drug Discov 2016; 11:907-16. [PMID: 27327499 DOI: 10.1080/17460441.2016.1201057] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION In the era of precision medicine and sophisticated modern genetics, the discovery of the BRAF(V600) inhibitor, vemurafenib, quickly became the model for targeted therapy in melanomas. As early as 2002, the majority of metastatic melanomas were described to harbor the BRAF(V600) mutation, setting the stage for an explosion of interest for targeting this protein as a novel therapeutic strategy. The highly selective BRAF(V600) inhibitor, vemurafenib, was identified initially through a large-scale drug screen. AREAS COVERED Here we examine vemurafenib's journey from discovery to clinical use in metastatic melanoma. Topics covered include preclinical data, single agent Phase 1,2 and 3 clinical trials, resistance issues and mechanisms, adverse effects including the development of squamous cell cancers, and combination trials. EXPERT OPINION Due to its tolerance, low toxicity profile, rapid tumor response, and improved outcomes in melanoma patients with BRAF(V600) mutations, vemurafenib was advanced rapidly through clinical trials to receive FDA approval in 2011. While its efficacy is well documented, durability has become an issue for most patients who experience therapeutic resistance in approximately 6-8 months. In addition, a concerning toxicity observed in patients taking the drug include development of localized cutaneous squamous cell carcinomas (SCCs). It is hypothesized that drug resistance and SCC development result from a similar paradoxical activation of protein signaling pathways, specifically MAPK. Identification of these mechanisms has led to additional treatment strategies involving new combination therapies.
Collapse
Affiliation(s)
- Alex Kim
- a Department of Surgery , University of Michigan , Ann Arbor , MI , USA
| | - Mark S Cohen
- a Department of Surgery , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
46
|
Medina TM, Lewis KD. The evolution of combined molecular targeted therapies to advance the therapeutic efficacy in melanoma: a highlight of vemurafenib and cobimetinib. Onco Targets Ther 2016; 9:3739-52. [PMID: 27382311 PMCID: PMC4922803 DOI: 10.2147/ott.s86774] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Metastatic melanoma is an aggressive, rapidly progressive disease which historically had very few effective treatment options. However, since 2011, the therapeutic landscape of melanoma has undergone a dramatic transformation with two distinct approaches and has catalyzed the successful advancement in the clinical field of immuno-oncology. In addition, the recognition of a key oncogenic driver mutation in melanoma, BRAF, stimulated the development of multiple potent kinase inhibitors which has also influenced the expansion and use of targeted agents in the practice of oncology. Vemurafenib, the initial BRAF inhibitor approved for the treatment of melanoma, was the first agent to demonstrate rapid clinical responses and significantly improved survival which was a clinical breakthrough in the treatment of melanoma. Although exciting and practice changing, the unparalleled responses with vemurafenib are usually not sustained. Further investigations delineated several mechanisms of acquired resistance which are most often mediated by the upregulation of the MAPK pathway. MEK inhibitors, another class of small-molecule inhibitors, were developed as an alternative agent to suppress the MAPK pathway downstream, independent from BRAF activation. Multiple studies have demonstrated the improvement in antitumor activity when MEK inhibitors are used in combination with BRAF inhibitors in the treatment of metastatic melanoma. This is a review of the investigations that led to the US Food and Drug Administration approval in 2015 of the combination of vemurafenib and cobimetinib, adding to the quickly growing armament for the treatment of advanced or metastatic melanoma with a BRAF V600 mutation.
Collapse
Affiliation(s)
- Theresa M Medina
- Cutaneous Oncology Program, Division of Medical Oncology, School of Medicine, University of Colorado – Anschutz Medical Campus, Aurora, CO, USA
| | - Karl D Lewis
- Cutaneous Oncology Program, Division of Medical Oncology, School of Medicine, University of Colorado – Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
47
|
Abstract
INTRODUCTION Since 2010 multiple targeted therapies and immunotherapies have been approved for the treatment of advanced melanoma. Pembrolizumab, a humanized monoclonal antibody directed against programed death receptor 1 has shown significant activity in advanced melanoma resulting in its approval first as post-ipilimumab and subsequently as frontline treatment. AREAS COVERED This article reviews the approved agents for the treatment of advanced melanoma with a focus on the preclinical and clinical evidence for the use of pembrolizumab in this setting. Primary emphasis is given to the clinical development of pembrolizumab, including phase I-III trials. Finally, we explore the role of pembrolizumab in combination with other therapies and ongoing investigations into its effectiveness in expanded patient populations. EXPERT OPINION Pembrolizumab provides durable responses and represents a major advancement in the treatment options for patients with advanced melanoma. Early studies of pembrolizumab in combination with other therapeutic agents have generated significant interest and further investigations including advanced clinical trials are warranted to evaluate safety and potential improved outcomes. Pembrolizumab and other immune checkpoint inhibitors are likely to play an expanded role in the treatment of advanced melanoma and other solid tumors over the next decade.
Collapse
Affiliation(s)
- Michael C Burns
- Vanderbilt University School of Medicine, Department of Medicine, Nashville, TN, USA
| | - Aidan O'Donnell
- Vanderbilt University School of Medicine, Department of Medicine, Nashville, TN, USA
| | - Igor Puzanov
- Vanderbilt-Ingram Cancer Center; Vanderbilt University Medical Center, Division of Hematology-Oncology, Nashville, TN, USA
| |
Collapse
|
48
|
Harris SJ, Brown J, Lopez J, Yap TA. Immuno-oncology combinations: raising the tail of the survival curve. Cancer Biol Med 2016; 13:171-93. [PMID: 27458526 PMCID: PMC4944548 DOI: 10.20892/j.issn.2095-3941.2016.0015] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/11/2016] [Indexed: 12/13/2022] Open
Abstract
There have been exponential gains in immuno-oncology in recent times through the development of immune checkpoint inhibitors. Already approved by the U.S. Food and Drug Administration for advanced melanoma and non-small cell lung cancer, immune checkpoint inhibitors also appear to have significant antitumor activity in multiple other tumor types. An exciting component of immunotherapy is the durability of antitumor responses observed, with some patients achieving disease control for many years. Nevertheless, not all patients benefit, and efforts should thus now focus on improving the efficacy of immunotherapy through the use of combination approaches and predictive biomarkers of response and resistance. There are multiple potential rational combinations using an immunotherapy backbone, including existing treatments such as radiotherapy, chemotherapy or molecularly targeted agents, as well as other immunotherapeutics. The aim of such antitumor strategies will be to raise the tail on the survival curve by increasing the number of long term survivors, while managing any additive or synergistic toxicities that may arise with immunotherapy combinations. Rational trial designs based on a clear understanding of tumor biology and drug pharmacology remain paramount. This article reviews the biology underpinning immuno-oncology, discusses existing and novel immunotherapeutic combinations currently in development, the challenges of predictive biomarkers of response and resistance and the impact of immuno-oncology on early phase clinical trial design.
Collapse
Affiliation(s)
| | | | | | - Timothy A. Yap
- Drug Development Unit
- Lung Unit, Royal Marsden Hospital and The Institute of Cancer Research, London SM2 5PT, UK
| |
Collapse
|
49
|
Busam KJ, Villain RE, Lum T, Busam JA, Hollmann TJ, P.M.Saw R, Coit DC, Scolyer RA, Wiesner T. Primary and Metastatic Cutaneous Melanomas Express ALK Through Alternative Transcriptional Initiation. Am J Surg Pathol 2016; 40:786-95. [PMID: 26872010 PMCID: PMC5037961 DOI: 10.1097/pas.0000000000000611] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A number of common driver mutations have been identified in melanoma, but other genetic or epigenetic aberrations are also likely to play a role in the pathogenesis of melanoma and present potential therapeutic targets. Translocations of the anaplastic lymphoma kinase (ALK), for example, have been reported in spitzoid melanocytic neoplasms leading to kinase-fusion proteins that result in immunohistochemically detectable ALK expression. In this study, we sought to determine whether ALK was also expressed in nonspitzoid primary and metastatic cutaneous melanomas. ALK immunohistochemistry was performed on 603 melanomas (303 primary and 300 metastatic tumors) from 600 patients. ALK immunohistochemistry expression was identified in 7 primary and 9 metastatic tumors. In 5 of 7 primary tumors and in 6 of 9 metastatic lesions, the majority of tumor cells were immunoreactive for ALK. In the other 2 primary and 3 metastatic lesions, positive staining was identified in less than half of the tumor cells. ALK positivity was found in the presence or absence of BRAF or NRAS mutations. In contrast to prior observations with ALK-positive Spitz tumors, none of the ALK-positive melanomas harbored a translocation. Instead, the ALK-positive melanomas predominantly expressed the recently described ALK isoform, ALK, which lacks the extracellular and transmembrane domains of wild-type ALK, consists primarily of the intracellular tyrosine kinase domain, and originates from an alternative transcriptional initiation site within the ALK gene. The findings are clinically relevant as patients with metastatic melanoma who have ALK expression may potentially benefit from treatment with ALK kinase inhibitors.
Collapse
Affiliation(s)
- Klaus J. Busam
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ricardo E. Villain
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
- Melanoma Institute of Australia, Sydney, Australia
- Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Trina Lum
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | | | - Travis J. Hollmann
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robyn P.M.Saw
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
- Melanoma Institute of Australia, Sydney, Australia
- Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Daniel C. Coit
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Richard A. Scolyer
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
- Melanoma Institute of Australia, Sydney, Australia
- Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Thomas Wiesner
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
50
|
Wellbrock C, Arozarena I. The Complexity of the ERK/MAP-Kinase Pathway and the Treatment of Melanoma Skin Cancer. Front Cell Dev Biol 2016; 4:33. [PMID: 27200346 PMCID: PMC4846800 DOI: 10.3389/fcell.2016.00033] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/12/2016] [Indexed: 12/17/2022] Open
Abstract
The central role played by the ERK/MAPK pathway downstream of RAS in human neoplasias is best exemplified in the context of melanoma skin cancer. Signaling through the MAPK pathway is crucial for the proliferation of melanocytes, the healthy pigment cells that give rise to melanoma. However, hyper-activation of the MAPK-pathway is found in over 90% of melanomas with approximately 50% of all patients displaying mutations in the kinase BRAF, and approximately 28% of all patients harboring mutations in the MAPK-pathway up-stream regulator NRAS. This finding has led to the development of BRAF and MEK inhibitors whose application in the clinic has shown unprecedented survival responses. Unfortunately the responses to MAPK pathway inhibitors are transient with most patients progressing within a year and a median progression free survival of 7-10 months. The disease progression is due to the development of drug-resistance based on various mechanisms, many of them involving a rewiring of the MAPK pathway. In this article we will review the complexity of MAPK signaling in melanocytic cells as well as the mechanisms of action of different MAPK-pathway inhibitors and their correlation with clinical response. We will reflect on mechanisms of innate and acquired resistance that limit patient's response, with a focus on the MAPK signaling network. Because of the resurgence of antibody-based immune-therapies there is a growing feeling of failure in the targeted therapy camp. However, recent studies have revealed new windows of therapeutic opportunity for melanoma sufferers treated with drugs targeting the MAPK pathway, and these opportunities will be discussed.
Collapse
Affiliation(s)
- Claudia Wellbrock
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of ManchesterManchester, UK
| | - Imanol Arozarena
- School of Applied Sciences, University of HuddersfieldHuddersfield, UK
| |
Collapse
|