1
|
Valentini E, Di Martile M, Brignone M, Di Caprio M, Manni I, Chiappa M, Sergio I, Chiacchiarini M, Bazzichetto C, Conciatori F, D'Aguanno S, D'Angelo C, Ragno R, Russillo M, Colotti G, Marchesi F, Bellone ML, Dal Piaz F, Felli MP, Damia G, Del Bufalo D. Bcl-2 family inhibitors sensitize human cancer models to therapy. Cell Death Dis 2023; 14:441. [PMID: 37460459 DOI: 10.1038/s41419-023-05963-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023]
Abstract
BH3 mimetics, targeting the Bcl-2 family anti-apoptotic proteins, represent a promising therapeutic opportunity in cancers. ABT-199, the first specific Bcl-2 inhibitor, was approved by FDA for the treatment of several hematological malignancies. We have recently discovered IS21, a novel pan BH3 mimetic with preclinical antitumor activity in several tumor types. Here, we evaluated the efficacy of IS21 and other BH3 mimetics, both as single agents and combined with the currently used antineoplastic agents in T-cell acute lymphoblastic leukemia, ovarian cancer, and melanoma. IS21 was found to be active in T-cell acute lymphoblastic leukemia, melanoma, lung, pancreatic, and ovarian cancer cell lines. Bcl-xL and Mcl-1 protein levels predicted IS21 sensitivity in melanoma and ovarian cancer, respectively. Exploring IS21 mechanism of action, we found that IS21 activity depends on the presence of BAX and BAK proteins: complexes between Bcl-2 and Bcl-xL proteins and their main binding partners were reduced after IS21 treatment. In combination experiments, BH3 mimetics sensitized leukemia cells to chemotherapy, ovarian cancer cells and melanoma models to PARP and MAPK inhibitors, respectively. We showed that this enhancing effect was related to the potentiation of the apoptotic pathway, both in hematologic and solid tumors. In conclusion, our data suggest the use of inhibitors of anti-apoptotic proteins as a therapeutic strategy to enhance the efficacy of anticancer treatment.
Collapse
Affiliation(s)
- Elisabetta Valentini
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Matteo Brignone
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marica Di Caprio
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Manni
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Michela Chiappa
- Laboratory of Gynecological Preclinical Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Sergio
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Chiacchiarini
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Chiara Bazzichetto
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabiana Conciatori
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Simona D'Aguanno
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carmen D'Angelo
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Rino Ragno
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza University of Rome, Rome, Italy
| | - Michelangelo Russillo
- Division of Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council, Rome, Italy
| | - Francesco Marchesi
- Hematology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Laura Bellone
- Department of Medicine, Surgery and Dentistry, University of Salerno, Fisciano, Italy
| | - Fabrizio Dal Piaz
- Department of Medicine, Surgery and Dentistry, University of Salerno, Fisciano, Italy
| | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanna Damia
- Laboratory of Gynecological Preclinical Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
2
|
Peng Z, Gillissen B, Richter A, Sinnberg T, Schlaak MS, Eberle J. Enhanced Apoptosis and Loss of Cell Viability in Melanoma Cells by Combined Inhibition of ERK and Mcl-1 Is Related to Loss of Mitochondrial Membrane Potential, Caspase Activation and Upregulation of Proapoptotic Bcl-2 Proteins. Int J Mol Sci 2023; 24:ijms24054961. [PMID: 36902392 PMCID: PMC10002974 DOI: 10.3390/ijms24054961] [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: 01/18/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Targeting of MAP kinase pathways by BRAF inhibitors has evolved as a key therapy for BRAF-mutated melanoma. However, it cannot be applied for BRAF-WT melanoma, and also, in BRAF-mutated melanoma, tumor relapse often follows after an initial phase of tumor regression. Inhibition of MAP kinase pathways downstream at ERK1/2, or inhibitors of antiapoptotic Bcl-2 proteins, such as Mcl-1, may serve as alternative strategies. As shown here, the BRAF inhibitor vemurafenib and the ERK inhibitor SCH772984 showed only limited efficacy in melanoma cell lines, when applied alone. However, in combination with the Mcl-1 inhibitor S63845, the effects of vemurafenib were strongly enhanced in BRAF-mutated cell lines, and the effects of SCH772984 were enhanced in both BRAF-mutated and BRAF-WT cells. This resulted in up to 90% loss of cell viability and cell proliferation, as well as in induction of apoptosis in up to 60% of cells. The combination of SCH772984/S63845 resulted in caspase activation, processing of poly (ADP-ribose) polymerase (PARP), phosphorylation of histone H2AX, loss of mitochondrial membrane potential, and cytochrome c release. Proving the critical role of caspases, a pan-caspase inhibitor suppressed apoptosis induction, as well as loss of cell viability. As concerning Bcl-2 family proteins, SCH772984 enhanced expression of the proapoptotic Bim and Puma, as well as decreased phosphorylation of Bad. The combination finally resulted in downregulation of antiapoptotic Bcl-2 and enhanced expression of the proapoptotic Noxa. In conclusion, combined inhibition of ERK and Mcl-1 revealed an impressive efficacy both in BRAF-mutated and WT melanoma cells, and may thus represent a new strategy for overcoming drug resistance.
Collapse
Affiliation(s)
- Zhe Peng
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Clinical Medicine, University of South China, Hengyang 421001, China
| | - Bernhard Gillissen
- Department of Hematology, Oncology, and Tumor Immunology, Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Antje Richter
- Department of Hematology, Oncology, and Tumor Immunology, Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Tobias Sinnberg
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Division of Dermatooncology, Department of Dermatology, University Tübingen, 72076 Tübingen, Germany
| | - Max S. Schlaak
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Eberle
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Correspondence:
| |
Collapse
|
3
|
Zhang ZY, Ding Y, Ezhilarasan R, Lhakhang T, Wang Q, Yang J, Modrek AS, Zhang H, Tsirigos A, Futreal A, Draetta GF, Verhaak RGW, Sulman EP. Lineage-coupled clonal capture identifies clonal evolution mechanisms and vulnerabilities of BRAF V600E inhibition resistance in melanoma. Cell Discov 2022; 8:102. [PMID: 36202798 PMCID: PMC9537441 DOI: 10.1038/s41421-022-00462-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
Targeted cancer therapies have revolutionized treatment but their efficacies are limited by the development of resistance driven by clonal evolution within tumors. We developed "CAPTURE", a single-cell barcoding approach to comprehensively trace clonal dynamics and capture live lineage-coupled resistant cells for in-depth multi-omics analysis and functional exploration. We demonstrate that heterogeneous clones, either preexisting or emerging from drug-tolerant persister cells, dominated resistance to vemurafenib in BRAFV600E melanoma. Further integrative studies uncovered diverse resistance mechanisms. This includes a previously unrecognized and clinically relevant mechanism, chromosome 18q21 gain, which leads to vulnerability of the cells to BCL2 inhibitor. We also identified targetable common dependencies of captured resistant clones, such as oxidative phosphorylation and E2F pathways. Our study provides new therapeutic insights into overcoming therapy resistance in BRAFV600E melanoma and presents a platform for exploring clonal evolution dynamics and vulnerabilities that can be applied to study treatment resistance in other cancers.
Collapse
Affiliation(s)
- Ze-Yan Zhang
- Department of Radiation Oncology, New York University (NYU) Grossman School of Medicine, New York, NY, USA.
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| | - Yingwen Ding
- Department of Radiation Oncology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Ravesanker Ezhilarasan
- Department of Radiation Oncology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Tenzin Lhakhang
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, NY, USA
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing, Jiangsu, China
| | - Jie Yang
- Department of Radiation Oncology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Aram S Modrek
- Department of Radiation Oncology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Hua Zhang
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, NY, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roel G W Verhaak
- Department of Computational Biology, The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Erik P Sulman
- Department of Radiation Oncology, New York University (NYU) Grossman School of Medicine, New York, NY, USA.
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
4
|
Simbulan-Rosenthal CM, Haribabu Y, Vakili S, Kuo LW, Clark H, Dougherty R, Alobaidi R, Carney B, Sykora P, Rosenthal DS. Employing CRISPR-Cas9 to Generate CD133 Synthetic Lethal Melanoma Stem Cells. Int J Mol Sci 2022; 23:2333. [PMID: 35216449 PMCID: PMC8877091 DOI: 10.3390/ijms23042333] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/19/2022] Open
Abstract
Malignant melanoma is a lethal skin cancer containing melanoma-initiating cells (MIC) implicated in tumorigenesis, invasion, and drug resistance, and is characterized by the elevated expression of stem cell markers, including CD133. The siRNA knockdown of CD133 enhances apoptosis induced by the MEK inhibitor trametinib in melanoma cells. This study investigates the underlying mechanisms of CD133's anti-apoptotic activity in patient-derived BAKP and POT cells, harboring difficult-to-treat NRASQ61K and NRASQ61R drivers, after CRISPR-Cas9 CD133 knockout or Dox-inducible expression of CD133. MACS-sorted CD133(+) BAKP cells were conditionally reprogrammed to derive BAKR cells with sustained CD133 expression and MIC features. Compared to BAKP, CD133(+) BAKR exhibit increased cell survival and reduced apoptosis in response to trametinib or the chemotherapeutic dacarbazine (DTIC). CRISPR-Cas9-mediated CD133 knockout in BAKR cells (BAKR-KO) re-sensitized cells to trametinib. CD133 knockout in BAKP and POT cells increased trametinib-induced apoptosis by reducing anti-apoptotic BCL-xL, p-AKT, and p-BAD and increasing pro-apoptotic BAX. Conversely, Dox-induced CD133 expression diminished apoptosis in both trametinib-treated cell lines, coincident with elevated p-AKT, p-BAD, BCL-2, and BCL-xL and decreased activation of BAX and caspases-3 and -9. AKT1/2 siRNA knockdown or inhibition of BCL-2 family members with navitoclax (ABT-263) in BAKP-KO cells further enhanced caspase-mediated apoptotic PARP cleavage. CD133 may therefore activate a survival pathway where (1) increased AKT phosphorylation and activation induces (2) BAD phosphorylation and inactivation, (3) decreases BAX activation, and (4) reduces caspases-3 and -9 activity and caspase-mediated PARP cleavage, leading to apoptosis suppression and drug resistance in melanoma. Targeting nodes of the CD133, AKT, or BCL-2 survival pathways with trametinib highlights the potential for combination therapies for NRAS-mutant melanoma stem cells for the development of more effective treatments for patients with high-risk melanoma.
Collapse
Affiliation(s)
- Cynthia M. Simbulan-Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Yogameenakshi Haribabu
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Sahar Vakili
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Li-Wei Kuo
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Havens Clark
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Ryan Dougherty
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Ryyan Alobaidi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Bonnie Carney
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
- Firefighters’ Burn and Surgical Laboratory, MedStar Health Research Institute, Washington, DC 20010, USA
| | - Peter Sykora
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
- Amelia Technologies, LLC, 1121 5th St. NW, Washington, DC 20001, USA
| | - Dean S. Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| |
Collapse
|
5
|
Coricovac D, Dehelean CA, Pinzaru I, Mioc A, Aburel OM, Macasoi I, Draghici GA, Petean C, Soica C, Boruga M, Vlaicu B, Muntean MD. Assessment of Betulinic Acid Cytotoxicity and Mitochondrial Metabolism Impairment in a Human Melanoma Cell Line. Int J Mol Sci 2021; 22:ijms22094870. [PMID: 34064489 PMCID: PMC8125295 DOI: 10.3390/ijms22094870] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/26/2021] [Accepted: 05/01/2021] [Indexed: 11/30/2022] Open
Abstract
Melanoma represents one of the most aggressive and drug resistant skin cancers with poor prognosis in its advanced stages. Despite the increasing number of targeted therapies, novel approaches are needed to counteract both therapeutic resistance and the side effects of classic therapy. Betulinic acid (BA) is a bioactive phytocompound that has been reported to induce apoptosis in several types of cancers including melanomas; however, its effects on mitochondrial bioenergetics are less investigated. The present study performed in A375 human melanoma cells was aimed to characterize the effects of BA on mitochondrial bioenergetics and cellular behavior. BA demonstrated a dose-dependent inhibitory effect in both mitochondrial respiration and glycolysis in A375 melanoma cells and at sub-toxic concentrations (10 μM) induced mitochondrial dysfunction by eliciting a decrease in the mitochondrial membrane potential and changes in mitochondria morphology and localization. In addition, BA triggered a dose-dependent cytotoxic effect characterized by apoptotic features: morphological alterations (nuclear fragmentation, apoptotic bodies) and the upregulation of pro-apoptotic markers mRNA expression (Bax, Bad and Bak). BA represents a viable therapeutic option via a complex modulatory effect on mitochondrial metabolism that might be useful in advanced melanoma or as reliable strategy to counteract resistance to standard therapy.
Collapse
Affiliation(s)
- Dorina Coricovac
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Cristina Adriana Dehelean
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Iulia Pinzaru
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
- Correspondence: (I.P.); (A.M.); Tel.: +40-256-494-604
| | - Alexandra Mioc
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
- Correspondence: (I.P.); (A.M.); Tel.: +40-256-494-604
| | - Oana-Maria Aburel
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
- Center for Translational Research and Systems Medicine, Faculty of Medicine,” Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq. no. 2, RO-300041 Timișoara, Romania
| | - Ioana Macasoi
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - George Andrei Draghici
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Crina Petean
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
| | - Codruta Soica
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Madalina Boruga
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
| | - Brigitha Vlaicu
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
| | - Mirela Danina Muntean
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
- Center for Translational Research and Systems Medicine, Faculty of Medicine,” Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq. no. 2, RO-300041 Timișoara, Romania
| |
Collapse
|
6
|
Trisciuoglio D, Del Bufalo D. New insights into the roles of antiapoptotic members of the Bcl-2 family in melanoma progression and therapy. Drug Discov Today 2021; 26:1126-1135. [PMID: 33545382 DOI: 10.1016/j.drudis.2021.01.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/25/2020] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
Prosurvival and antiapoptotic B cell lymphoma-2 (Bcl-2) family proteins are often overexpressed in cutaneous melanoma, one of the most aggressive types of human cancer. They are also implicated in resistance to therapy and participate in melanoma progression by regulating various processes, including cell proliferation, migration, invasion, and crosstalk with the tumor microenvironment. In this review, we summarize recent findings related to prosurvival members of the Bcl-2 family beyond their canonical functions in the apoptotic pathway, mainly focusing on their potential roles as diagnostic and prognostic biomarkers in cutaneous melanoma. We also provide an overview of different approaches used to inhibit Bcl-2 proteins in preclinical and clinical studies, which are mainly based on the inhibition of protein expression or the disruption of their antiapoptotic functions.
Collapse
Affiliation(s)
- Daniela Trisciuoglio
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome, Italy; Institute of Molecular Biology and Pathology, National Research Council, via degli Apuli 4, 00185, Rome, Italy.
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome, Italy.
| |
Collapse
|
7
|
Abstract
About half of all cutaneous melanomas harbor activating mutations in the BRAF oncogene. Dependence on this pathway makes the tumors vulnerable to BRAF (and downstream MEK) inhibition, and three drug combinations are approved to target this vulnerability in advanced melanomas with BRAFV600 mutations. Responses to BRAF/MEK inhibitors are usually fast, but durability of response can be limited. Five-year data from BRAF/MEK inhibitors show long-term survival benefit for a third of the patients. There is a wide variety of known mechanisms of resistance to BRAF/MEK inhibition, such as mitogen-activated protein kinase reactivation, activation of parallel pathways, alterations in cell-cycle regulation, and non-genetic resistance mechanisms. Strategies that have been explored to overcome these mechanisms include alternative dosing regimens, addition of another kinase inhibitor, and use of anti-PD-1 immunotherapy either in combination or post-relapse on BRAF/MEK inhibitor therapies.
Collapse
|
8
|
Hartman ML, Gajos-Michniewicz A, Talaj JA, Mielczarek-Lewandowska A, Czyz M. BH3 mimetics potentiate pro-apoptotic activity of encorafenib in BRAF V600E melanoma cells. Cancer Lett 2020; 499:122-136. [PMID: 33259900 DOI: 10.1016/j.canlet.2020.11.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 12/29/2022]
Abstract
BRAFV600- and MEK1/2-targeting therapies rarely produce durable response in melanoma patients. We investigated five BRAFV600E melanoma cell lines derived from drug-naïve tumor specimens to assess cell death response to encorafenib (Braftovi), a recently FDA-approved BRAFV600 inhibitor. Drug-naïve cell lines (i) did not harbor damaging alterations in genes encoding core apoptotic machinery, but they differed in (ii) mitochondrial priming as demonstrated by whole-cell BH3 profiling, and (iii) levels of selected anti-apoptotic proteins. Encorafenib modulated the balance between apoptosis-regulating proteins as it upregulated BIM and BMF, and attenuated NOXA, but did not affect the levels of pro-survival proteins except for MCL-1 and BCL-XL in selected cell lines. Induction of apoptosis could be predicted using Dynamic BH3 profiling. The extent of apoptosis was dependent on both (i) cell-intrinsic proximity to the apoptotic threshold (initial mitochondrial priming) and (ii) the abundance of encorafenib-induced BIM (iBIM; drug-induced change in priming). While co-inhibition of MCL-1 and BCL-XL/BCL-2 was indispensable for apoptosis in drug-naïve cells, encorafenib altered cell dependence to MCL-1, and reliance on BCL-XL/BCL-2 was additionally found in cell lines that were highly primed to apoptosis by encorafenib. This translated into robust apoptosis when encorafenib was combined with selective BH3 mimetics. Our study provides a mechanistic insight into the role of proteins from the BCL-2 family in melanoma cell response to targeted therapy, and presents preclinical evidence that (i) MCL-1 is a druggable target to potentiate encorafenib activity, whereas (ii) pharmacological inhibition of BCL-XL/BCL-2 might be relevant but only for a narrow group of encorafenib-treated patients.
Collapse
Affiliation(s)
- Mariusz L Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland.
| | - Anna Gajos-Michniewicz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| | - Julita A Talaj
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| | | | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| |
Collapse
|
9
|
Proietti I, Skroza N, Bernardini N, Tolino E, Balduzzi V, Marchesiello A, Michelini S, Volpe S, Mambrin A, Mangino G, Romeo G, Maddalena P, Rees C, Potenza C. Mechanisms of Acquired BRAF Inhibitor Resistance in Melanoma: A Systematic Review. Cancers (Basel) 2020; 12:E2801. [PMID: 33003483 PMCID: PMC7600801 DOI: 10.3390/cancers12102801] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
This systematic review investigated the literature on acquired v-raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitor resistance in patients with melanoma. We searched MEDLINE for articles on BRAF inhibitor resistance in patients with melanoma published since January 2010 in the following areas: (1) genetic basis of resistance; (2) epigenetic and transcriptomic mechanisms; (3) influence of the immune system on resistance development; and (4) combination therapy to overcome resistance. Common resistance mutations in melanoma are BRAF splice variants, BRAF amplification, neuroblastoma RAS viral oncogene homolog (NRAS) mutations and mitogen-activated protein kinase kinase 1/2 (MEK1/2) mutations. Genetic and epigenetic changes reactivate previously blocked mitogen-activated protein kinase (MAPK) pathways, activate alternative signaling pathways, and cause epithelial-to-mesenchymal transition. Once BRAF inhibitor resistance develops, the tumor microenvironment reverts to a low immunogenic state secondary to the induction of programmed cell death ligand-1. Combining a BRAF inhibitor with a MEK inhibitor delays resistance development and increases duration of response. Multiple other combinations based on known mechanisms of resistance are being investigated. BRAF inhibitor-resistant cells develop a range of 'escape routes', so multiple different treatment targets will probably be required to overcome resistance. In the future, it may be possible to personalize combination therapy towards the specific resistance pathway in individual patients.
Collapse
Affiliation(s)
- Ilaria Proietti
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Nevena Skroza
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Nicoletta Bernardini
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Ersilia Tolino
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Veronica Balduzzi
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Anna Marchesiello
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Simone Michelini
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Salvatore Volpe
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Alessandra Mambrin
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Giorgio Mangino
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (G.R.)
| | - Giovanna Romeo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (G.R.)
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, 00185 Rome, Italy
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00185 Rome, Italy
| | - Patrizia Maddalena
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | | | - Concetta Potenza
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| |
Collapse
|
10
|
Mukherjee N, Skees J, Todd KJ, West DA, Lambert KA, Robinson WA, Amato CM, Couts KL, Van Gulick R, MacBeth M, Nassar K, Tan AC, Zhai Z, Fujita M, Bagby SM, Dart CR, Lambert JR, Norris DA, Shellman YG. MCL1 inhibitors S63845/MIK665 plus Navitoclax synergistically kill difficult-to-treat melanoma cells. Cell Death Dis 2020; 11:443. [PMID: 32513939 PMCID: PMC7280535 DOI: 10.1038/s41419-020-2646-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023]
Abstract
Current treatment for patients with metastatic melanoma include molecular-targeted therapies and immune checkpoint inhibitors. However, a subset of melanomas are difficult-to-treat. These melanomas include those without the genetic markers for targeted therapy, non-responsive to immunotherapy, and those who have relapsed or exhausted their therapeutic options. Therefore, it is necessary to understand and explore other biological processes that may provide new therapeutic approaches. One of most appealing is targeting the apoptotic/anti-apoptotic system that is effective against leukemia. We used genetic knockdown and pharmacologic approaches of BH3 mimetics to target anti-apoptotic BCL2 family members and identified MCL1 and BCLXL as crucial pro-survival members in melanoma. We then examined the effects of combining BH3 mimetics to target MCL1 and BCLXL in vitro and in vivo. These include clinical-trial-ready compounds such as ABT-263 (Navitoclax) and S63845/S64315 (MIK655). We used cell lines derived from patients with difficult-to-treat melanomas. In vitro, the combined inhibition of MCL1 and BCLXL resulted in significantly effective cell killing compared to single-agent treatment (p < 0.05) in multiple assays, including sphere assays. The combination-induced cell death was independent of BIM, and NOXA. Recapitulated in our mouse xenograft model, the combination inhibited tumor growth, reduced sphere-forming capacity (p < 0.01 and 0.05, respectively), and had tolerable toxicity (p > 0.40). Taken together, this study suggests that dual targeting of MCL1 and BCLXL should be considered as a treatment option for difficult-to-treat melanoma patients.
Collapse
Affiliation(s)
- Nabanita Mukherjee
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
| | - Jenette Skees
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
| | - Kaleb J Todd
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
| | - Drake A West
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
| | - Karoline A Lambert
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
| | - William A Robinson
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - Carol M Amato
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - Kasey L Couts
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - Robert Van Gulick
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - Morgan MacBeth
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - Kelsey Nassar
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - Aik-Choon Tan
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, US
| | - Zili Zhai
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
| | - Stacey M Bagby
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - Chiara R Dart
- University of Colorado Anschutz Medical Campus, School of Medicine, Division of Medical Oncology, Mail Stop 8117, Aurora, CO, 80045, US
| | - James R Lambert
- Department of Pathology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8104, Aurora, CO, 80045, US
| | - David A Norris
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US
- Department of Veterans Affairs Medical Center, Dermatology Section, Denver, CO, 80220, US
| | - Yiqun G Shellman
- Department of Dermatology, University of Colorado Anschutz Medical Campus, School of Medicine, Mail Stop 8127, Aurora, CO, 80045, US.
- University of Colorado Anschutz Medical Campus, Gates Center for Regenerative Medicine, Aurora, CO, 80045, US.
| |
Collapse
|
11
|
Montero J, Gstalder C, Kim DJ, Sadowicz D, Miles W, Manos M, Cidado JR, Paul Secrist J, Tron AE, Flaherty K, Stephen Hodi F, Yoon CH, Letai A, Fisher DE, Haq R. Destabilization of NOXA mRNA as a common resistance mechanism to targeted therapies. Nat Commun 2019; 10:5157. [PMID: 31727958 PMCID: PMC6856172 DOI: 10.1038/s41467-019-12477-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 08/06/2019] [Indexed: 12/15/2022] Open
Abstract
Most targeted cancer therapies fail to achieve complete tumor regressions or attain durable remissions. To understand why these treatments fail to induce robust cytotoxic responses despite appropriately targeting oncogenic drivers, here we systematically interrogated the dependence of cancer cells on the BCL-2 family of apoptotic proteins after drug treatment. We observe that multiple targeted therapies, including BRAF or EGFR inhibitors, rapidly deplete the pro-apoptotic factor NOXA, thus creating a dependence on the anti-apoptotic protein MCL-1. This adaptation requires a pathway leading to destabilization of the NOXA mRNA transcript. We find that interruption of this mechanism of anti-apoptotic adaptive resistance dramatically increases cytotoxic responses in cell lines and a murine melanoma model. These results identify NOXA mRNA destabilization/MCL-1 adaptation as a non-genomic mechanism that limits apoptotic responses, suggesting that sequencing of MCL-1 inhibitors with targeted therapies could overcome such widespread and clinically important resistance. MAPK-targeted therapies fail to achieve complete remission. Here, the authors show that anti-apoptosis resistance is acquired in these targeted therapies through the mRNA destabilization of NOXA which leads to dependence on MCL-1, and that sequential combination of MCL-1 inhibition with targeted therapies overcomes this resistance.
Collapse
Affiliation(s)
- Joan Montero
- Division of Hematologic Neoplasia/Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA.,Institute for Bioengineering of Catalonia, C/Baldiri Reixac 15-21, Ed. Hèlix 3ª planta · 08028, Barcelona, Spain
| | - Cécile Gstalder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA.,Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA
| | - Daniel J Kim
- Department of Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, 44 Fruit Street, Boston, MA, 02114, USA
| | - Dorota Sadowicz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA.,Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA
| | - Wayne Miles
- Department of Molecular Genetics, The Ohio State University, 820 Biomedical Research Tower 460 West 12th Avenue, Columbus, 43210, OH, USA
| | - Michael Manos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA
| | - Justin R Cidado
- Bioscience, Oncology IMED Biotech Unit, AstraZeneca, 35 Gatehouse Dr, Waltham, Boston, 02451, MA, USA
| | - J Paul Secrist
- Bioscience, Oncology IMED Biotech Unit, AstraZeneca, 35 Gatehouse Dr, Waltham, Boston, 02451, MA, USA.,LifeMine Therapeutics, 100 Acorn Park Drive, 6th Floor Cambridge, Cambridge, MA, 02140, USA
| | - Adriana E Tron
- Bioscience, Oncology IMED Biotech Unit, AstraZeneca, 35 Gatehouse Dr, Waltham, Boston, 02451, MA, USA
| | - Keith Flaherty
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, 44 Fruit Street, Boston, MA, 02114, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA
| | - Charles H Yoon
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, 02115, USA
| | - Anthony Letai
- Division of Hematologic Neoplasia/Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA
| | - David E Fisher
- Department of Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, 44 Fruit Street, Boston, MA, 02114, USA. .,Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, 44 Fruit Street, Boston, MA, 02114, USA.
| | - Rizwan Haq
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA. .,Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, 02115, MA, USA.
| |
Collapse
|
12
|
Sale MJ, Minihane E, Monks NR, Gilley R, Richards FM, Schifferli KP, Andersen CL, Davies EJ, Vicente MA, Ozono E, Markovets A, Dry JR, Drew L, Flemington V, Proia T, Jodrell DI, Smith PD, Cook SJ. Targeting melanoma's MCL1 bias unleashes the apoptotic potential of BRAF and ERK1/2 pathway inhibitors. Nat Commun 2019; 10:5167. [PMID: 31727888 PMCID: PMC6856071 DOI: 10.1038/s41467-019-12409-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 09/06/2019] [Indexed: 01/01/2023] Open
Abstract
BRAF and MEK1/2 inhibitors are effective in melanoma but resistance inevitably develops. Despite increasing the abundance of pro-apoptotic BIM and BMF, ERK1/2 pathway inhibition is predominantly cytostatic, reflecting residual pro-survival BCL2 family activity. Here, we show that uniquely low BCL-XL expression in melanoma biases the pro-survival pool towards MCL1. Consequently, BRAF or MEK1/2 inhibitors are synthetic lethal with the MCL1 inhibitor AZD5991, driving profound tumour cell death that requires BAK/BAX, BIM and BMF, and inhibiting tumour growth in vivo. Combination of ERK1/2 pathway inhibitors with BCL2/BCL-w/BCL-XL inhibitors is stronger in CRC, correlating with a low MCL1:BCL-XL ratio; indeed the MCL1:BCL-XL ratio is predictive of ERK1/2 pathway inhibitor synergy with MCL1 or BCL2/BCL-w/BCL-XL inhibitors. Finally, AZD5991 delays acquired BRAFi/MEKi resistance and enhances the efficacy of an ERK1/2 inhibitor in a model of acquired BRAFi + MEKi resistance. Thus combining ERK1/2 pathway inhibitors with MCL1 antagonists in melanoma could improve therapeutic index and patient outcomes.
Collapse
Affiliation(s)
- Matthew J Sale
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - Emma Minihane
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Noel R Monks
- Oncology R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Rebecca Gilley
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Frances M Richards
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Kevin P Schifferli
- Oncology R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | | | - Emma J Davies
- Oncology R&D, AstraZeneca, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Mario Aladren Vicente
- CRUK Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Eiko Ozono
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | | | - Jonathan R Dry
- Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, MA, 02451, USA
| | - Lisa Drew
- Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, MA, 02451, USA
| | - Vikki Flemington
- Oncology R&D, AstraZeneca, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Theresa Proia
- Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, MA, 02451, USA
| | - Duncan I Jodrell
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Paul D Smith
- Oncology R&D, AstraZeneca, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Simon J Cook
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
13
|
Fujimura T, Fujisawa Y, Kambayashi Y, Aiba S. Significance of BRAF Kinase Inhibitors for Melanoma Treatment: From Bench to Bedside. Cancers (Basel) 2019; 11:cancers11091342. [PMID: 31514399 PMCID: PMC6770075 DOI: 10.3390/cancers11091342] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/09/2019] [Indexed: 01/10/2023] Open
Abstract
According to clinical trials, BRAF kinase inhibitors in combination with MEK kinase inhibitors are among the most promising chemotherapy regimens for the treatment of advanced BRAF-mutant melanoma, though the rate of BRAF mutation gene-bearing cutaneous melanoma is limited, especially in the Asian population. In addition, drug resistance sometimes abrogates the persistent efficacy of combined therapy with BRAF and MEK inhibitors. Therefore, recent pre-clinical study-based clinical trials have attempted to identify optimal drugs (e.g., immune checkpoint inhibitors or histone deacetylase (HDAC) inhibitors) that improve the anti-melanoma effects of BRAF and MEK inhibitors. In addition, the development of novel protocols to avoid resistance of BRAF inhibitors is another purpose of recent pre-clinical and early clinical trials. This review focuses on pre-clinical studies and early to phase III clinical trials to discuss the development of combined therapy based on BRAF inhibitors for BRAF-mutant advanced melanoma, as well as mechanisms of resistance to BRAF inhibitors.
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.
| | - Yasuhiro Fujisawa
- Department of Dermatology, University of Tsukuba, Tsukuba 305-8576, Japan.
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.
| |
Collapse
|
14
|
Lim B, Greer Y, Lipkowitz S, Takebe N. Novel Apoptosis-Inducing Agents for the Treatment of Cancer, a New Arsenal in the Toolbox. Cancers (Basel) 2019; 11:cancers11081087. [PMID: 31370269 PMCID: PMC6721450 DOI: 10.3390/cancers11081087] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Evasion from apoptosis is an important hallmark of cancer cells. Alterations of apoptosis pathways are especially critical as they confer resistance to conventional anti-cancer therapeutics, e.g., chemotherapy, radiotherapy, and targeted therapeutics. Thus, successful induction of apoptosis using novel therapeutics may be a key strategy for preventing recurrence and metastasis. Inhibitors of anti-apoptotic molecules and enhancers of pro-apoptotic molecules are being actively developed for hematologic malignancies and solid tumors in particular over the last decade. However, due to the complicated apoptosis process caused by a multifaceted connection with cross-talk pathways, protein–protein interaction, and diverse resistance mechanisms, drug development within the category has been extremely challenging. Careful design and development of clinical trials incorporating predictive biomarkers along with novel apoptosis-inducing agents based on rational combination strategies are needed to ensure the successful development of these molecules. Here, we review the landscape of currently available direct apoptosis-targeting agents in clinical development for cancer treatment and update the related biomarker advancement to detect and validate the efficacy of apoptosis-targeted therapies, along with strategies to combine them with other agents.
Collapse
Affiliation(s)
- Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yoshimi Greer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Naoko Takebe
- Early Clinical Trials Development, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
15
|
Rice SJ, Liu X, Wang HG, Belani CP. EGFR mutations and AKT phosphorylation are markers for sensitivity to combined MCL-1 and BCL-2/xL inhibition in non-small cell lung cancer. PLoS One 2019; 14:e0217657. [PMID: 31150457 PMCID: PMC6544263 DOI: 10.1371/journal.pone.0217657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is among the common and deadly cancers. Although the treatment options for late-stage cancer patients have continued to increase in numbers, the overall survival rates for these patients have not shown significant improvement. This highlights the need for new targets and drugs to more effectively treat lung cancer patients. In this study, we characterize the MCL-1 inhibitor maritoclax alone or in combination with a BCL-2/xL inhibitor in a panel of lung cancer cell lines. BCL-2 family proteins, phosphorylated proteins, and apoptosis were monitored following the treatments. We found that maritoclax was effective at inhibiting growth in these lung cancer cells. We also establish that cell lines with EGFR mutations were most sensitive to the combined inhibition of MCL-1 and BCL-2/xL. In addition, a high level of phosphorylated AKT (S473) was identified as a marker for sensitivity to the combination treatment. This work has defined EGFR mutations and AKT phosphorylation as markers for sensitivity to combined MCL-1 and BCL-2/xL targeted therapy and establishes a rationale to explore multiple BCL-2 family members in patients who are refractory to EGFR inhibitor treatment. Our data support the design of a clinical trial that aims to employ inhibitors of the BCL-2 family of proteins in lung cancer patients.
Collapse
Affiliation(s)
- Shawn J. Rice
- Penn State Hershey Cancer Institute, Hershey, Pennsylvania, United States of America
| | - Xin Liu
- Penn State Hershey Cancer Institute, Hershey, Pennsylvania, United States of America
| | - Hong-Gang Wang
- Penn State Hershey Cancer Institute, Hershey, Pennsylvania, United States of America
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Chandra P. Belani
- Penn State Hershey Cancer Institute, Hershey, Pennsylvania, United States of America
- Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Jeong JH, Oh JM, Jeong SY, Lee SW, Lee J, Ahn BC. Combination Treatment with the BRAF V600E Inhibitor Vemurafenib and the BH3 Mimetic Navitoclax for BRAF-Mutant Thyroid Carcinoma. Thyroid 2019; 29:540-548. [PMID: 30869573 DOI: 10.1089/thy.2018.0511] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Vemurafenib is a selective BRAF inhibitor (BRAFi) that has shown promising activity in BRAFV600E-positive papillary thyroid cancer (PTC). However, adverse events and resistance to a single-agent BRAFi often require discontinuation of the targeted therapy in BRAFV600E-positive PTC. Thus, this study investigated the expression of anti-apoptotic B-cell lymphoma 2 (BCL-2) family members, which are frequently overexpressed in many human cancers to inhibit apoptosis, in PTC harboring the BRAFV600E mutation after BRAFi treatment, and then evaluated the cytotoxic effects of a homology 3 domain (BH3)-mimetic in combination with a BRAFi. METHODS K1 cells (BRAFV600E-positive human PTC) were treated with various concentrations of vemurafenib to investigate the effect of the BRAFi. In addition, the study analyzed the protein expression profiles of phosphorylated ERK1/2 (p-ERK 1/2) and anti-apoptotic BCL-2 family after vemurafenib treatment and selected the target anti-apoptotic protein. Antitumor effects were measured by cell counting, and effects on apoptosis were determined by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay and Western blot analysis. RESULTS At a concentration of 10 μM, vemurafenib inhibited the growth of K1 cells by 49.4%. Western blot analysis following exposure to 10 μM vemurafenib revealed that p-ERK1/2 gradually decreased over 24 hours, but the expression of B-cell lymphoma-extralarge (BCL-XL) and BCL-2 increased after 12 hours of treatment. Based on this result, the K1 cells were treated with navitoclax (BCL-2/BCL-XL inhibitor) for 24 hours up to a concentration of 4 μM, which resulted in negligible effects on cell survival. However, a combination treatment of 0.5 μM navitoclax with 1 μM vemurafenib resulted in significantly enhanced cell growth inhibition and increased apoptosis. CONCLUSIONS The results of the present study show that vemurafenib increased the expression of anti-apoptotic proteins of the BCL-2 family. Thus, the combination of vemurafenib with navitoclax may be effective in BRAFV600E-positive PTC treatment.
Collapse
Affiliation(s)
- Ju Hye Jeong
- 1 Department of Nuclear medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji Min Oh
- 2 Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Shin Young Jeong
- 1 Department of Nuclear medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
- 2 Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- 3 Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sang-Woo Lee
- 1 Department of Nuclear medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
- 2 Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- 3 Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jaetae Lee
- 2 Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- 3 Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- 2 Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- 3 Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
17
|
BH3 mimetics induce apoptosis independent of DRP-1 in melanoma. Cell Death Dis 2018; 9:907. [PMID: 30185782 PMCID: PMC6125485 DOI: 10.1038/s41419-018-0932-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/11/2018] [Accepted: 07/19/2018] [Indexed: 12/17/2022]
Abstract
Despite the recent advancement in treating melanoma, options are still limited for patients without BRAF mutations or in relapse from current treatments. BH3 mimetics against members of the BCL-2 family have gained excitement with the recent success in hematological malignancies. However, single drug BH3 mimetic therapy in melanoma has limited effectiveness due to escape by the anti-apoptotic protein MCL-1 and/or survival of melanoma-initiating cells (MICs). We tested the efficacy of the BH3 mimetic combination of A-1210477 (an MCL-1 inhibitor) and ABT-263 (a BCL-2/BCL-XL/BCL-W inhibitor) in killing melanoma, especially MICs. We also sought to better define Dynamin-Related Protein 1 (DRP-1)'s role in melanoma; DRP-1 is known to interact with members of the BCL-2 family and is a possible therapeutic target for melanoma treatment. We used multiple assays (cell viability, apoptosis, bright field, immunoblot, and sphere formation), as well as the CRISPR/Cas9 genome-editing techniques. For clinical relevance, we employed patient samples of different mutation status, including some relapsed from current treatments such as anti-PD-1 immunotherapy. We found the BH3 mimetic combination kill both the MICs and non-MICs (bulk of melanoma) in all cell lines and patient samples irrespective of the mutation status or relapsed state (p < 0.05). Unexpectedly, the major pro-apoptotic proteins, NOXA and BIM, are not necessary for the combination-induced cell death. Furthermore, the combination impedes the activation of DRP-1, and inhibition of DRP-1 further enhances apoptosis (p < 0.05). DRP-1 effects in melanoma differ from those seen in other cancer cells. These results provide new insights into BCL-2 family's regulation of the apoptotic pathway in melanoma, and suggest that inhibiting the major anti-apoptotic proteins is sufficient to induce cell death even without involvement from major pro-apoptotic proteins. Importantly, our study also indicates that DRP-1 inhibition is a promising adjuvant for BH3 mimetics in melanoma treatment.
Collapse
|
18
|
Abstract
Introduction Breakthroughs in targeted therapy have significantly improved outcomes for many patients with advanced melanoma, including those with BRAFV600 mutant disease. Targeted therapy for BRAFV600-mutant metastatic melanoma includes combinations of BRAF inhibitors and MEK inhibitors, which improve response rates and prolong progression-free survival (PFS) and overall survival (OS) in these patients. However, while durable responses have been observed, many patients develop acquired resistance to these drugs. Areas covered Recent clinical trial updates and ongoing studies with targeted therapy for BRAF-V600 mutant melanoma are reviewed. Expert opinion Although BRAF targeted therapy remains an effective treatment for BRAF-mutant for melanoma, ongoing trials are exploring combinations with other targeted therapeutics and immunotherapeutics to determine whether tumor responses can be prolonged, and these drugs are increasingly utilized in the neoadjuvant and adjuvant settings.
Collapse
Affiliation(s)
- Zeynep Eroglu
- The Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, 10920 McKinley Dr, Tampa, FL, USA
| | - Alpaslan Ozgun
- The Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, 10920 McKinley Dr, Tampa, FL, USA
| |
Collapse
|
19
|
Eroglu Z, Chen YA, Gibney GT, Weber JS, Kudchadkar RR, Khushalani NI, Markowitz J, Brohl AS, Tetteh LF, Ramadan H, Arnone G, Li J, Zhao X, Sharma R, Darville LNF, Fang B, Smalley I, Messina JL, Koomen JM, Sondak VK, Smalley KSM. Combined BRAF and HSP90 Inhibition in Patients with Unresectable BRAF V600E-Mutant Melanoma. Clin Cancer Res 2018; 24:5516-5524. [PMID: 29674508 DOI: 10.1158/1078-0432.ccr-18-0565] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 03/20/2018] [Accepted: 04/17/2018] [Indexed: 12/16/2022]
Abstract
Purpose: BRAF inhibitors are clinically active in patients with advanced BRAFV600-mutant melanoma, although acquired resistance remains common. Preclinical studies demonstrated that resistance could be overcome using concurrent treatment with the HSP90 inhibitor XL888.Patients and Methods: Vemurafenib (960 mg p.o. b.i.d.) combined with escalating doses of XL888 (30, 45, 90, or 135 mg p.o. twice weekly) was investigated in 21 patients with advanced BRAFV600-mutant melanoma. Primary endpoints were safety and determination of a maximum tolerated dose. Correlative proteomic studies were performed to confirm HSP inhibitor activity.Results: Objective responses were observed in 15 of 20 evaluable patients [75%; 95% confidence interval (CI), 51%-91%], with 3 complete and 12 partial responses. Median progression-free survival and overall survival were 9.2 months (95% CI, 3.8-not reached) and 34.6 months (6.2-not reached), respectively. The most common grade 3/4 toxicities were skin toxicities, such as rash (n = 4, 19%) and cutaneous squamous cell carcinomas (n = 3, 14%), along with diarrhea (n = 3, 14%). Pharmacodynamic analysis of patients' peripheral blood mononuclear cells (PBMC) showed increased day 8 HSP70 expression compared with baseline in the three cohorts with XL888 doses ≥45 mg. Diverse effects of vemurafenib-XL888 upon intratumoral HSP client protein expression were noted, with the expression of multiple proteins (including ERBB3 and BAD) modulated on therapy.Conclusions: XL888 in combination with vemurafenib has clinical activity in patients with advanced BRAFV600-mutant melanoma, with a tolerable side-effect profile. HSP90 inhibitors warrant further evaluation in combination with current standard-of-care BRAF plus MEK inhibitors in BRAFV600-mutant melanoma. Clin Cancer Res; 24(22); 5516-24. ©2018 AACR See related commentary by Sullivan, p. 5496.
Collapse
Affiliation(s)
- Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Y Ann Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Geoffrey T Gibney
- Georgetown Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | | | - Ragini R Kudchadkar
- Winship Cancer Institute of Emory University School of Medicine, Atlanta, Georgia
| | | | - Joseph Markowitz
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Andrew S Brohl
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Leticia F Tetteh
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Howida Ramadan
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Gina Arnone
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Jiannong Li
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Xiuhua Zhao
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Ritin Sharma
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, Florida
| | | | - Bin Fang
- Department of Proteomics, Moffitt Cancer Center, Tampa, Florida
| | - Inna Smalley
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, Florida
| | - Jane L Messina
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - John M Koomen
- Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Keiran S M Smalley
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida. .,Department of Tumor Biology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
20
|
Mukherjee N, Almeida A, Partyka KA, Lu Y, Schwan JV, Lambert K, Rogers M, Robinson WA, Robinson SE, Applegate AJ, Amato CM, Luo Y, Fujita M, Norris DA, Shellman YG. Combining a GSI and BCL-2 inhibitor to overcome melanoma's resistance to current treatments. Oncotarget 2018; 7:84594-84607. [PMID: 27829238 PMCID: PMC5356684 DOI: 10.18632/oncotarget.13141] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/27/2016] [Indexed: 12/14/2022] Open
Abstract
Major limitations of current melanoma treatments are for instances of relapse and the lack of therapeutic options for BRAF wild-type patients who do not respond to immunotherapy. Many studies therefore focus on killing resistant subpopulations, such as Melanoma Initiating Cells (MICs) to prevent relapse. Here we examined whether combining a GSI (γ-Secretase Inhibitor) with ABT-737 (a small molecule BCL-2/BCL-XL/BCL-W inhibitor) can kill both the non-MICs (bulk of melanoma) and MICs. To address the limitations of melanoma therapies, we included multiple tumor samples of patients relapsed from current treatments, with a diverse genetic background (with or without the common BRAF, NRAS or NF1 mutations) in these studies. Excitingly, the combination treatment reduced cell viability and induced apoptosis of the non-MICs; disrupted primary spheres, decreased the ALDH+ cells, and inhibited the self-renewability of the MICs in multiple melanoma cell lines and relapsed patient samples. Using a low-cell-number mouse xenograft model, we demonstrated that the combination significantly reduced the tumor initiating ability of MIC-enriched cultures from relapsed patient samples. Mechanistic studies also indicate that cell death is NOXA-dependent. In summary, this combination may be a promising strategy to address treatment relapse and for triple wild-type patients who do not respond to immunotherapy.
Collapse
Affiliation(s)
- Nabanita Mukherjee
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - Adam Almeida
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - Katie A Partyka
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - Yan Lu
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - Josianna V Schwan
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - Karoline Lambert
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - Madison Rogers
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - William A Robinson
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO 80045, USA
| | - Steven E Robinson
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO 80045, USA
| | - Allison J Applegate
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO 80045, USA
| | - Carol M Amato
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO 80045, USA
| | - Yuchun Luo
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - Mayumi Fujita
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| | - David A Norris
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA.,Department of Veterans Affairs Medical Center, Dermatology Section, Denver, CO 80220, USA
| | - Yiqun G Shellman
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology, Aurora, CO 80045, USA
| |
Collapse
|
21
|
Song KA, Niederst MJ, Lochmann TL, Hata AN, Kitai H, Ham J, Floros KV, Hicks MA, Hu H, Mulvey HE, Drier Y, Heisey DAR, Hughes MT, Patel NU, Lockerman EL, Garcia A, Gillepsie S, Archibald HL, Gomez-Caraballo M, Nulton TJ, Windle BE, Piotrowska Z, Sahingur SE, Taylor SM, Dozmorov M, Sequist LV, Bernstein B, Ebi H, Engelman JA, Faber AC. Epithelial-to-Mesenchymal Transition Antagonizes Response to Targeted Therapies in Lung Cancer by Suppressing BIM. Clin Cancer Res 2018; 24:197-208. [PMID: 29051323 PMCID: PMC5959009 DOI: 10.1158/1078-0432.ccr-17-1577] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/13/2017] [Accepted: 10/13/2017] [Indexed: 12/26/2022]
Abstract
Purpose: Epithelial-to-mesenchymal transition (EMT) confers resistance to a number of targeted therapies and chemotherapies. However, it has been unclear why EMT promotes resistance, thereby impairing progress to overcome it.Experimental Design: We have developed several models of EMT-mediated resistance to EGFR inhibitors (EGFRi) in EGFR-mutant lung cancers to evaluate a novel mechanism of EMT-mediated resistance.Results: We observed that mesenchymal EGFR-mutant lung cancers are resistant to EGFRi-induced apoptosis via insufficient expression of BIM, preventing cell death despite potent suppression of oncogenic signaling following EGFRi treatment. Mechanistically, we observed that the EMT transcription factor ZEB1 inhibits BIM expression by binding directly to the BIM promoter and repressing transcription. Derepression of BIM expression by depletion of ZEB1 or treatment with the BH3 mimetic ABT-263 to enhance "free" cellular BIM levels both led to resensitization of mesenchymal EGFR-mutant cancers to EGFRi. This relationship between EMT and loss of BIM is not restricted to EGFR-mutant lung cancers, as it was also observed in KRAS-mutant lung cancers and large datasets, including different cancer subtypes.Conclusions: Altogether, these data reveal a novel mechanistic link between EMT and resistance to lung cancer targeted therapies. Clin Cancer Res; 24(1); 197-208. ©2017 AACR.
Collapse
Affiliation(s)
- Kyung-A Song
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Matthew J Niederst
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Timothy L Lochmann
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Aaron N Hata
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hidenori Kitai
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Jungoh Ham
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Konstantinos V Floros
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Mark A Hicks
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Haichuan Hu
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hillary E Mulvey
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Yotam Drier
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Daniel A R Heisey
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Mark T Hughes
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Neha U Patel
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Elizabeth L Lockerman
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Angel Garcia
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Shawn Gillepsie
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hannah L Archibald
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Maria Gomez-Caraballo
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Tara J Nulton
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Brad E Windle
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Zofia Piotrowska
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Sinem E Sahingur
- Department of Periodontics, VCU School of Dentistry, Virginia Commonwealth University, Richmond, Virginia
| | - Shirley M Taylor
- Department of Microbiology and Immunology, Massey Cancer Center, Richmond, Virginia
| | - Mikhail Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Bradley Bernstein
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hiromichi Ebi
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Anthony C Faber
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia.
| |
Collapse
|
22
|
Abstract
Progress in understanding and treating metastatic melanoma is the result of decades of basic and translational research as well as the development of better in vitro tools for modeling the disease. Here, we review the latest therapeutic options for metastatic melanoma and the known genetic and non-genetic mechanisms of resistance to these therapies, as well as the in vitro toolbox that has provided the greatest insights into melanoma progression. These include next-generation sequencing technologies and more complex 2D and 3D cell culture models to functionally test the data generated by genomics approaches. The combination of hypothesis generating and hypothesis testing paradigms reviewed here will be the foundation for the next phase of metastatic melanoma therapies in the coming years.
Collapse
|
23
|
Abstract
OPINION STATEMENT Advanced melanoma is related to a very grim prognosis and fast progression. Until recently, there has been no indicated treatment that would affect the disease's outcome. However, the progress in immunotherapy and molecular therapy has significantly changed the unfavourable prognosis of melanoma progression and its short survival rate. Both approaches have improved patients' outcomes and provided renewed hope for successful treatment. Moreover, in order to further enhance patients' outcomes and to avoid mechanisms of tumour resistance, investigators attempted a combined approach. Targeted therapy combinations allowed a better response rate and progression-free survival than monotherapy with one of the agents. Another promising combination, but with limiting toxicities, is a concurrent immuno- and molecular-targeted therapy. It is suspected that complimentary usage of these drugs may lead to synergism, providing robust and quick tumour responses as well as long-lasting effects. Results of currently ongoing clinical trials that investigate combination strategies in melanoma are expected to provide more mature data about the effectiveness and the safety profile of those therapies. Until more robust results of these studies occur, the best management of advanced and metastatic melanoma is immunotherapy with anti-PD1 drugs or targeted therapy with concomitant BRAF and MEK inhibitor. However, which of these two options should be used first is still under discussion.
Collapse
|
24
|
Abstract
Treatment options for patients with metastatic melanoma, and especially BRAF-mutant melanoma, have changed dramatically in the past 5 years, with the FDA approval of eight new therapeutic agents. During this period, the treatment paradigm for BRAF-mutant disease has evolved rapidly: the standard-of-care BRAF-targeted approach has shifted from single-agent BRAF inhibition to combination therapy with a BRAF and a MEK inhibitor. Concurrently, immunotherapy has transitioned from cytokine-based treatment to antibody-mediated blockade of the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and, now, the programmed cell-death protein 1 (PD-1) immune checkpoints. These changes in the treatment landscape have dramatically improved patient outcomes, with the median overall survival of patients with advanced-stage melanoma increasing from approximately 9 months before 2011 to at least 2 years - and probably longer for those with BRAF-V600-mutant disease. Herein, we review the clinical trial data that established the standard-of-care treatment approaches for advanced-stage melanoma. Mechanisms of resistance and biomarkers of response to BRAF-targeted treatments and immunotherapies are discussed, and the contrasting clinical benefits and limitations of these therapies are explored. We summarize the state of the field and outline a rational approach to frontline-treatment selection for each individual patient with BRAF-mutant melanoma.
Collapse
|
25
|
Gambichler T, Rooms I, Scholl L, Stockfleth E, Stücker M, Sand M. BH3-only protein Bim predicts advanced stage of cutaneous melanoma. J Eur Acad Dermatol Venereol 2016; 30:1926-1929. [PMID: 27356803 DOI: 10.1111/jdv.13791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/06/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Bim having strong pro-apoptotic effects belongs to the BH3-only proteins of the Bcl-2 protein family and contributes to survival pathways in cancer cells. OBJECTIVES We aimed to investigate Bim protein expression in cutaneous melanoma (CM). METHODS Bim protein expression was assessed by immunohistochemistry in primary and metastatic melanomas and correlated with clinical and histopathological features. RESULTS The Bim immunoreactivity score of the primary melanomas investigated (4.6 ± 1.5) was significantly (P < 0.0001) higher than that observed in metastases (2.8 ± 1.1). Low Bim expression was significantly associated with primary nodular melanoma type (P = 0.005). Moreover, Bim expression was significantly inversely correlated with tumour thickness (r = -0.36; P = 0.0035), advanced stage of disease (stage III and IV; r = -0.60; P < 0.0001), disease relapse (r = -0.18; P = 0.034) and disease-related death (r = -0.19; P = 0.026). Advanced stage of disease was independently predicted by low Bim expression (P = 0.0010, odds ratio: 0.22, 95% CI: 0.10-0.56) on multivariate analysis; however, Bim was not shown to be an independent predictor for disease relapse (P = 0.40) and disease-related death (P = 0.77). CONCLUSIONS Our data demonstrate that Bim protein expression is significantly inversely correlated with melanoma features that are associated with worse prognosis. We have shown that Bim protein expression in CM is an independent predictor for advanced disease confirming that this pro-apoptotic BH3-only protein might be a potent biomarker and promising therapeutic target.
Collapse
Affiliation(s)
- T Gambichler
- Skin Cancer Center of the Department of Dermatology, Ruhr-University Bochum, Bochum, Germany.
| | - I Rooms
- Skin Cancer Center of the Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - L Scholl
- Skin Cancer Center of the Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - E Stockfleth
- Skin Cancer Center of the Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - M Stücker
- Skin Cancer Center of the Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - M Sand
- Skin Cancer Center of the Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
26
|
Kawakami H, Huang S, Pal K, Dutta SK, Mukhopadhyay D, Sinicrope FA. Mutant BRAF Upregulates MCL-1 to Confer Apoptosis Resistance that Is Reversed by MCL-1 Antagonism and Cobimetinib in Colorectal Cancer. Mol Cancer Ther 2016; 15:3015-3027. [PMID: 27765849 DOI: 10.1158/1535-7163.mct-16-0017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 08/26/2016] [Accepted: 09/27/2016] [Indexed: 01/19/2023]
Abstract
Oncogenic BRAFV600E mutations activate MAPK signaling and are associated with treatment resistance and poor prognosis in patients with colorectal cancer. In BRAFV600E-mutant colorectal cancers, treatment failure may be related to BRAFV600E-mediated apoptosis resistance that occurs by an as yet undefined mechanism. We found that BRAFV600E can upregulate anti-apoptotic MCL-1 in a gene dose-dependent manner using colorectal cancer cell lines isogenic for BRAF BRAFV600E-induced MCL-1 upregulation was confirmed by ectopic BRAFV600E expression that activated MEK/ERK signaling to phosphorylate (MCL-1Thr163) and stabilize MCL-1. Upregulation of MCL-1 was mediated by MEK/ERK shown by the ability of ERK siRNA to suppress MCL-1. Stabilization of MCL-1 by phosphorylation was shown by a phosphorylation-mimicking mutant and an unphosphorylated MCL-1 mutant that decreased or increased MCL-1 protein turnover, respectively. MEK/ERK inhibition by cobimetinib suppressed MCL-1 expression/phosphorylation and induced proapoptotic BIM to a greater extent than did vemurafenib in BRAFV600E cell lines. MCL-1 knockdown versus control shRNA significantly enhanced cobimetinib-induced apoptosis in vitro and in HT29 colon cancer xenografts. The small-molecule MCL-1 inhibitor, A-1210477, also enhanced cobimetinib-induced apoptosis in vitro that was due to disruption of the interaction of MCL-1 with proapoptotic BAK and BIM. Knockdown of BIM attenuated BAX, but not BAK, activation by cobimetinib plus A-1210477. In summary, BRAFV600E-mediated MEK/ERK activation can upregulate MCL-1 by phosphorylation/stabilization to confer apoptosis resistance that can be reversed by MCL-1 antagonism combined with cobimetinib, suggesting a novel therapeutic strategy against BRAFV600E-mutant CRCs. Mol Cancer Ther; 15(12); 3015-27. ©2016 AACR.
Collapse
Affiliation(s)
- Hisato Kawakami
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Shengbing Huang
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida
| | | | - Frank A Sinicrope
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
27
|
Fofaria NM, Frederick DT, Sullivan RJ, Flaherty KT, Srivastava SK. Overexpression of Mcl-1 confers resistance to BRAFV600E inhibitors alone and in combination with MEK1/2 inhibitors in melanoma. Oncotarget 2016; 6:40535-56. [PMID: 26497853 PMCID: PMC4747351 DOI: 10.18632/oncotarget.5755] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 09/20/2015] [Indexed: 01/13/2023] Open
Abstract
Melanoma harboring BRAF mutations frequently develop resistance to BRAF inhibitors, limiting the impact of treatment. Here, we establish a mechanism of resistance and subsequently identified a suitable drug combination to overcome the resistance. Single treatment of BRAF mutant melanoma cell lines with vemurafenib or dabrafenib (BRAF inhibitors) alone or in combination with trametinib (MEK1/2 inhibitor) resulted in overexpression of Mcl-1. Overexpression of Mcl-1 in A375 and SK-MEL-28 by transfection completely blocked BRAF and MEK1/2 inhibitor-mediated inhibition of cell survival and apoptosis. Melanoma cells resistant to BRAF inhibitors showed massive expression of Mcl-1 as compared to respective sensitive cell lines. Silencing of Mcl-1 using siRNA completely sensitized resistant melanoma cells to growth suppression and induction of apoptosis by BRAF inhibitors. In vivo, vemurafenib resistant A375 xenografts implanted in athymic nude mice showed substantial tumor growth inhibition when treated with a combination of vemurafenib and Mcl-1 inhibitor or siRNA. Immunohistochemistry and western blot analyses demonstrated enhanced expression of Mcl-1 and activation of ERK1/2 in vemurafenib-resistant tumors whereas level of Mcl-1 or p-ERK1/2 was diminished in the tumors of mice treated with either of the combination. Biopsied tumors from the patients treated with or resistant to BRAF inhibitors revealed overexpression of Mcl-1. These results suggest that the combination of BRAF inhibitors with Mcl-1 inhibitor may have therapeutic advantage to melanoma patients with acquired resistance to BRAF inhibitors alone or in combination with MEK1/2 inhibitors.
Collapse
Affiliation(s)
- Neel M Fofaria
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Dennie T Frederick
- Harvard Medical School, Boston, Massachusetts, USA.,Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Ryan J Sullivan
- Harvard Medical School, Boston, Massachusetts, USA.,Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Keith T Flaherty
- Harvard Medical School, Boston, Massachusetts, USA.,Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Sanjay K Srivastava
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| |
Collapse
|
28
|
Emmons MF, Faião-Flores F, Smalley KSM. The role of phenotypic plasticity in the escape of cancer cells from targeted therapy. Biochem Pharmacol 2016; 122:1-9. [PMID: 27349985 DOI: 10.1016/j.bcp.2016.06.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/23/2016] [Indexed: 01/01/2023]
Abstract
Targeted therapy has proven to be beneficial at producing significant responses in patients with a wide variety of cancers. Despite initially impressive responses, most individuals ultimately fail these therapies and show signs of drug resistance. Very few patients are ever cured. Emerging evidence suggests that treatment of cancer cells with kinase inhibitors leads a minor population of cells to undergo a phenotypic switch to a more embryonic-like state. The adoption of this state, which is analogous to an epithelial-to-mesenchymal transition, is associated with drug resistance and increased tumor aggressiveness. In this commentary we will provide a comprehensive analysis of the mechanisms that underlie the embryonic reversion that occurs on targeted cancer therapy and will review potential novel therapeutic strategies designed to eradicate the escaping cells.
Collapse
Affiliation(s)
- Michael F Emmons
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Fernanda Faião-Flores
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA; The Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Keiran S M Smalley
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA; The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA.
| |
Collapse
|
29
|
Akabane H, Sullivan RJ. The Future of Molecular Analysis in Melanoma: Diagnostics to Direct Molecularly Targeted Therapy. Am J Clin Dermatol 2016; 17:1-10. [PMID: 26518880 DOI: 10.1007/s40257-015-0159-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Melanoma is a malignancy of pigment-producing cells that is driven by a variety of genetic mutations and aberrations. In most cases, this leads to upregulation of the mitogen-activated protein kinase (MAPK) pathway through activating mutations of upstream mediators of the pathway including BRAF and NRAS. With the advent of effective MAPK pathway inhibitors, including the US FDA-approved BRAF inhibitors vemurafenib and dabrafenib and MEK inhibitor trametinib, molecular analysis has become an integral part of the care of patients with metastatic melanoma. In this article, the key molecular targets and strategies to inhibit these targets therapeutically are presented, and the techniques of identifying these targets, in both tissue and blood, are discussed.
Collapse
Affiliation(s)
- Hugo Akabane
- Department of Medicine, Metrowest Medical Center, Framingham, MA, USA
| | - Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
30
|
Karachaliou N, Pilotto S, Teixidó C, Viteri S, González-Cao M, Riso A, Morales-Espinosa D, Molina MA, Chaib I, Santarpia M, Richardet E, Bria E, Rosell R. Melanoma: oncogenic drivers and the immune system. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:265. [PMID: 26605311 PMCID: PMC4630557 DOI: 10.3978/j.issn.2305-5839.2015.08.06] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 12/19/2022]
Abstract
Advances and in-depth understanding of the biology of melanoma over the past 30 years have contributed to a change in the consideration of melanoma as one of the most therapy-resistant malignancies. The finding that oncogenic BRAF mutations drive tumor growth in up to 50% of melanomas led to a molecular therapy revolution for unresectable and metastatic disease. Moving beyond BRAF, inactivation of immune regulatory checkpoints that limit T cell responses to melanoma has provided targets for cancer immunotherapy. In this review, we discuss the molecular biology of melanoma and we focus on the recent advances of molecularly targeted and immunotherapeutic approaches.
Collapse
|
31
|
Abstract
We describe the landscape of genomic alterations in cutaneous melanomas through DNA, RNA, and protein-based analysis of 333 primary and/or metastatic melanomas from 331 patients. We establish a framework for genomic classification into one of four subtypes based on the pattern of the most prevalent significantly mutated genes: mutant BRAF, mutant RAS, mutant NF1, and Triple-WT (wild-type). Integrative analysis reveals enrichment of KIT mutations and focal amplifications and complex structural rearrangements as a feature of the Triple-WT subtype. We found no significant outcome correlation with genomic classification, but samples assigned a transcriptomic subclass enriched for immune gene expression associated with lymphocyte infiltrate on pathology review and high LCK protein expression, a T cell marker, were associated with improved patient survival. This clinicopathological and multi-dimensional analysis suggests that the prognosis of melanoma patients with regional metastases is influenced by tumor stroma immunobiology, offering insights to further personalize therapeutic decision-making.
Collapse
Affiliation(s)
- The Cancer Genome Atlas Network
- Cancer Genome Atlas Program Office, National Cancer Institute at NIH, 31 Center Drive, Bldg. 31, Suite 3A20, Bethesda, MD 20892, USA
| |
Collapse
|
32
|
Sullivan RJ, Flaherty KT. New Strategies in Melanoma: Entering the Era of Combinatorial Therapy. Clin Cancer Res 2015; 21:2424-35. [DOI: 10.1158/1078-0432.ccr-14-1650] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Hata AN, Engelman JA, Faber AC. The BCL2 Family: Key Mediators of the Apoptotic Response to Targeted Anticancer Therapeutics. Cancer Discov 2015; 5:475-87. [PMID: 25895919 DOI: 10.1158/2159-8290.cd-15-0011] [Citation(s) in RCA: 446] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/27/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED The ability of cancer cells to suppress apoptosis is critical for carcinogenesis. The BCL2 family proteins comprise the sentinel network that regulates the mitochondrial or intrinsic apoptotic response. Recent advances in our understanding of apoptotic signaling pathways have enabled methods to identify cancers that are "primed" to undergo apoptosis, and have revealed potential biomarkers that may predict which cancers will undergo apoptosis in response to specific therapies. Complementary efforts have focused on developing novel drugs that directly target antiapoptotic BCL2 family proteins. In this review, we summarize the current knowledge of the role of BCL2 family members in cancer development and response to therapy, focusing on targeted therapeutics, recent progress in the development of apoptotic biomarkers, and therapeutic strategies designed to overcome deficiencies in apoptosis. SIGNIFICANCE Apoptosis, long known to be important for response to conventional cytotoxic chemotherapy, has more recently been shown to be essential for the efficacy of targeted therapies. Approaches that increase the likelihood of a cancer to undergo apoptosis following therapy may help improve targeted treatment strategies. Cancer Discov; 5(5); 475-87. ©2015 AACR.
Collapse
Affiliation(s)
- Aaron N Hata
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Anthony C Faber
- Virginia Commonwealth University Philips Institute for Oral Health Research, School of Dentistry and Massey Cancer Center, Richmond, Virginia.
| |
Collapse
|
34
|
The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat Genet 2015; 47:250-6. [PMID: 25665005 DOI: 10.1038/ng.3218] [Citation(s) in RCA: 419] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 01/15/2015] [Indexed: 12/20/2022]
Abstract
Resistance to RAF- and MEK-targeted therapy is a major clinical challenge. RAF and MEK inhibitors are initially but only transiently effective in some but not all patients with BRAF gene mutation and are largely ineffective in those with RAS gene mutation because of resistance. Through a genetic screen in BRAF-mutant tumor cells, we show that the Hippo pathway effector YAP (encoded by YAP1) acts as a parallel survival input to promote resistance to RAF and MEK inhibitor therapy. Combined YAP and RAF or MEK inhibition was synthetically lethal not only in several BRAF-mutant tumor types but also in RAS-mutant tumors. Increased YAP in tumors harboring BRAF V600E was a biomarker of worse initial response to RAF and MEK inhibition in patients, establishing the clinical relevance of our findings. Our data identify YAP as a new mechanism of resistance to RAF- and MEK-targeted therapy. The findings unveil the synthetic lethality of combined suppression of YAP and RAF or MEK as a promising strategy to enhance treatment response and patient survival.
Collapse
|
35
|
Sullivan R, LoRusso P, Boerner S, Dummer R. Achievements and challenges of molecular targeted therapy in melanoma. Am Soc Clin Oncol Educ Book 2015:177-186. [PMID: 25993155 DOI: 10.14694/edbook_am.2015.35.177] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The treatment of melanoma has been revolutionized over the past decade with the development of effective molecular and immune targeted therapies. The great majority of patients with melanoma have mutations in oncogenes that predominantly drive signaling through the mitogen activated protein kinase (MAPK) pathway. Analytic tools have been developed that can effectively stratify patients into molecular subsets based on the identification of mutations in oncogenes and/or tumor suppressor genes that drive the MAPK pathway. At the same time, potent and selective inhibitors of mediators of the MAPK pathway such as RAF, MEK, and ERK have become available. The most dramatic example is the development of single-agent inhibitors of BRAF (vemurafenib, dabrafenib, encorafenib) and MEK (trametinib, cobimetinib, binimetinib) for patients with metastatic BRAFV600-mutant melanoma, a subset that represents 40% to 50% of patients with metastatic melanoma. More recently, the elucidation of mechanisms underlying resistance to single-agent BRAF inhibitor therapy led to a second generation of trials that demonstrated the superiority of BRAF inhibitor/MEK inhibitor combinations (dabrafenib/trametinib; vemurafenib/cobimetinib) compared to single-agent BRAF inhibitors. Moving beyond BRAFV600 targeting, a number of other molecular subsets--such as mutations in MEK, NRAS, and non-V600 BRAF and loss of function of the tumor suppressor neurofibromatosis 1 (NF1)--are predicted to respond to MAPK pathway targeting by single-agent pan-RAF, MEK, or ERK inhibitors. As these strategies are being tested in clinical trials, preclinical and early clinical trial data are now emerging about which combinatorial approaches might be best for these patients.
Collapse
Affiliation(s)
- Ryan Sullivan
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Patricia LoRusso
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Scott Boerner
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Awad MM, Sullivan RJ. Dabrafenib in combination with trametinib for the treatment of metastatic melanoma. Expert Rev Clin Pharmacol 2014; 8:25-33. [DOI: 10.1586/17512433.2015.974556] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Ascierto PA, Grimaldi AM, Anderson AC, Bifulco C, Cochran A, Garbe C, Eggermont AM, Faries M, Ferrone S, Gershenwald JE, Gajewski TF, Halaban R, Hodi FS, Kefford R, Kirkwood JM, Larkin J, Leachman S, Maio M, Marais R, Masucci G, Melero I, Palmieri G, Puzanov I, Ribas A, Saenger Y, Schilling B, Seliger B, Stroncek D, Sullivan R, Testori A, Wang E, Ciliberto G, Mozzillo N, Marincola FM, Thurin M. Future perspectives in melanoma research: meeting report from the "Melanoma Bridge", Napoli, December 5th-8th 2013. J Transl Med 2014; 12:277. [PMID: 25348889 PMCID: PMC4232645 DOI: 10.1186/s12967-014-0277-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 09/23/2014] [Indexed: 12/28/2022] Open
Abstract
The fourth "Melanoma Bridge Meeting" took place in Naples, December 5 to 8th, 2013. The four topics discussed at this meeting were: Diagnosis and New Procedures, Molecular Advances and Combination Therapies, News in Immunotherapy, and Tumor Microenvironment and Biomarkers.
Collapse
Affiliation(s)
- Paolo A Ascierto
- />Istituto Nazionale Tumori, Fondazione “G. Pascale”, Napoli, Italy
| | | | | | - Carlo Bifulco
- />Translational Molecular Pathology, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
| | - Alistair Cochran
- />Departments of Pathology and Laboratory Medicine and Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), John Wayne Cancer Institute, Santa Monica, CA USA
| | - Claus Garbe
- />Center for Dermato Oncology, Department of Dermatology, University of Tübingen, Tübingen, Germany
| | | | - Mark Faries
- />Donald L. Morton Melanoma Research Program, John Wayne Cancer Institute, Santa Monica, CA USA
| | - Soldano Ferrone
- />Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Jeffrey E Gershenwald
- />Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Thomas F Gajewski
- />Departments of Medicine and of Pathology, Immunology and Cancer Program, The University of Chicago Medicine, Chicago, IL USA
| | - Ruth Halaban
- />Department of Dermatology, Yale University School of Medicine, New Haven, CT USA
| | - F Stephen Hodi
- />Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Richard Kefford
- />Westmead Institute for Cancer Research, Westmead Millennium Institute and Melanoma Institute Australia, University of Sydney, Sydney, NSW Australia
| | - John M Kirkwood
- />Division of Hematology/Oncology, Departments of Medicine, Dermatology, and Translational Science, University of Pittsburgh School of Medicine and Melanoma Program of the Pittsburgh Cancer Institute, Pittsburgh, PA USA
| | - James Larkin
- />Royal Marsden NHS Foundation Trust, London, UK
| | - Sancy Leachman
- />Department of Dermatology, Oregon Health Sciences University, Portland, OR USA
| | - Michele Maio
- />Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Richard Marais
- />Molecular Oncology Group, The Paterson Institute for Cancer Research, Wilmslow Road, Manchester, M20 4BX UK
| | - Giuseppe Masucci
- />Department of Oncology-Pathology, The Karolinska Hospital, Stockholm, Sweden
| | - Ignacio Melero
- />Centro de Investigación Médica Aplicada, Clinica Universidad de Navarra, Pamplona, Navarra Spain
| | - Giuseppe Palmieri
- />Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, Sassari, Italy
| | - Igor Puzanov
- />Vanderbilt University Medical Center, Nashville, TN USA
| | - Antoni Ribas
- />Tumor Immunology Program, Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA USA
| | - Yvonne Saenger
- />Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Bastian Schilling
- />Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- />German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Barbara Seliger
- />Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Halle, Germany
| | - David Stroncek
- />Cell Processing Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD USA
| | - Ryan Sullivan
- />Center for Melanoma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA USA
| | | | - Ena Wang
- />Division Chief of Translational Medicine, Sidra Medical and Research Centre, Doha, Qatar
| | | | - Nicola Mozzillo
- />Istituto Nazionale Tumori, Fondazione “G. Pascale”, Napoli, Italy
| | | | - Magdalena Thurin
- />Cancer Diagnosis Program, National Cancer Institute, NIH, Bethesda, MD USA
| |
Collapse
|