1
|
Xue Y, Chen T, Ma Z, Pu X, Xu J, Zhai S, Du X, Ji Y, Simon MC, Zhai W, Xue W. Osalmid sensitizes clear cell Renal Cell Carcinoma to navitoclax through a STAT3/BCL-XL pathway. Cancer Lett 2025:217514. [PMID: 39894195 DOI: 10.1016/j.canlet.2025.217514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common and lethal urinary malignancy characterized by its resistance to apoptosis. Despite the emerging treatment options available for ccRCC, only a small proportion of patients achieve long-term survival benefits. Previous studies have demonstrated that inducing tumor cell senescence, followed by treatment using senolytics, represents a potential strategy for triggering tumor cell apoptosis. However, it remains unclear whether this strategy is suitable for the treatment of ccRCC. Using the whole-genome CRISPR screening database Dependency Map portal (DepMap), we identified ribonucleotide reductase family member 2 (RRM2), which catalyzes the conversion of ribonucleotides to deoxyribonucleotides (dNTPs), as an essential targetable gene for ccRCC. Herein, we report that the combination of the choleretic drug osalmid targeting RRM2 and the senolytic compound navitoclax targeting BCL-XL represents a novel therapeutic approach for ccRCC. Furthermore, we have validated this approach across a panel of human ccRCC cells with different genetic backgrounds and multiple preclinical models, including cell line-derived xenografts (CDX), patient-derived xenografts (PDX), and patient-derived organoids (PDO). Mechanistically, osalmid-mediated inhibition of dNTPs generation induces cellular senescence in ccRCC, concomitant with STAT3 activation and upregulation of BCL-XL, thus rendering these cells vulnerable to navitoclax, which targets the BCL-2 protein family.
Collapse
Affiliation(s)
- Yizheng Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Tianyi Chen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| | - Zehua Ma
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550001, China
| | - Xinyuan Pu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| | - Junyao Xu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| | - Shuanfeng Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| | - Yiyi Ji
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA, Howard Hughes Medical Institute
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Wei Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Immune Therapy Institute State, Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127,China.
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
2
|
Markouli M, Pagoni MN, Diamantopoulos P. BCL-2 inhibitors in hematological malignancies: biomarkers that predict response and management strategies. Front Oncol 2025; 14:1501950. [PMID: 39906657 PMCID: PMC11790632 DOI: 10.3389/fonc.2024.1501950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Apoptosis is an essential characteristic of cancer and its dysregular promotes tumor growth, clonal evolution, and treatment resistance. B-cell lymphoma-2 (BCL-2) protein family members are key to the intrinsic, mitochondrial apoptotic pathway. The inhibition of the BCL-2 family pro-survival proteins, which are frequently overexpressed in B-cell malignancies and pose a fundamental carcinogenic mechanism has been proposed as a promising therapeutic option, with venetoclax (ABT-199) being the first FDA-approved BCL-2 inhibitor. Unfortunately, although BCL-2 inhibition has shown remarkable results in a range of B-cell lymphoid cancers as well as acute myeloid leukemia (AML), the development of resistance significantly reduces response rates in specific tumor subtypes. In this article, we explain the role of BCL-2 family proteins in apoptosis and their mechanism of action that justifies their inhibition as a potential treatment target in B-cell malignancies, including chronic lymphocytic leukemia, multiple myeloma, B-cell lymphomas, but also AML. We further analyze the tumor characteristics that result in the development of intrinsic or inherited resistance to BCL-2 inhibitors. Finally, we focus on the biomarkers that can be used to predict responses to treatment in the name of personalized medicine, with the goal of exploring alternative strategies to overcome resistance.
Collapse
Affiliation(s)
- Mariam Markouli
- Department of Internal Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| | - Maria N. Pagoni
- Department of Hematology-Lymphomas and BMT Unit, Evangelismos Hospital, Athens, Greece
| | - Panagiotis Diamantopoulos
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
3
|
Fernandez EG, Mai WX, Song K, Bayley NA, Kim J, Zhu H, Pioso M, Young P, Andrasz CL, Cadet D, Liau LM, Li G, Yong WH, Rodriguez FJ, Dixon SJ, Souers AJ, Li JJ, Graeber TG, Cloughesy TF, Nathanson DA. Integrated molecular and functional characterization of the intrinsic apoptotic machinery identifies therapeutic vulnerabilities in glioma. Nat Commun 2024; 15:10089. [PMID: 39572533 PMCID: PMC11582606 DOI: 10.1038/s41467-024-54138-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/03/2024] [Indexed: 11/24/2024] Open
Abstract
Genomic profiling often fails to predict therapeutic outcomes in cancer. This failure is, in part, due to a myriad of genetic alterations and the plasticity of cancer signaling networks. Functional profiling, which ascertains signaling dynamics, is an alternative method to anticipate drug responses. It is unclear whether integrating genomic and functional features of solid tumours can provide unique insight into therapeutic vulnerabilities. We perform combined molecular and functional characterization, via BH3 profiling of the intrinsic apoptotic machinery, in glioma patient samples and derivative models. We identify that standard-of-care therapy rapidly rewires apoptotic signaling in a genotype-specific manner, revealing targetable apoptotic vulnerabilities in gliomas containing specific molecular features (e.g., TP53 WT). However, integration of BH3 profiling reveals high mitochondrial priming is also required to induce glioma apoptosis. Accordingly, a machine-learning approach identifies a composite molecular and functional signature that best predicts responses of diverse intracranial glioma models to standard-of-care therapies combined with ABBV-155, a clinical drug targeting intrinsic apoptosis. This work demonstrates how complementary functional and molecular data can robustly predict therapy-induced cell death.
Collapse
Affiliation(s)
- Elizabeth G Fernandez
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Wilson X Mai
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kai Song
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nicholas A Bayley
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Jiyoon Kim
- Department of Biostatistics, Jonathan and Karin Fielding School of Public Health, Los Angeles, California, USA
| | - Henan Zhu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Marissa Pioso
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Pauline Young
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Cassidy L Andrasz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles, CA, 90095-7088, USA
| | - Dimitri Cadet
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Linda M Liau
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gang Li
- Department of Biostatistics, Jonathan and Karin Fielding School of Public Health, Los Angeles, California, USA
| | - William H Yong
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Fausto J Rodriguez
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Andrew J Souers
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Jingyi Jessica Li
- Department of Biostatistics, Jonathan and Karin Fielding School of Public Health, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, 90095-7088, USA
- Department of Computational Medicine, University of California, Los Angeles, CA, 90095-1766, USA
- Department of Statistics and Data Science, University of California, Los Angeles, CA, 90095-1554, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- UCLA Metabolomics Center, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy F Cloughesy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Ji Y, Harris MA, Newton LM, Harris TJ, Fairlie WD, Lee EF, Hawkins CJ. Osteosarcoma cells depend on MCL-1 for survival, and osteosarcoma metastases respond to MCL-1 antagonism plus regorafenib in vivo. BMC Cancer 2024; 24:1350. [PMID: 39497108 PMCID: PMC11533409 DOI: 10.1186/s12885-024-13088-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
Osteosarcoma is the most common form of primary bone cancer, which primarily afflicts children and adolescents. Chemotherapy, consisting of doxorubicin, cisplatin and methotrexate (MAP) increased the 5-year osteosarcoma survival rate from 20% to approximately 60% by the 1980s. However, osteosarcoma survival rates have remained stagnant for several decades. Patients whose disease fails to respond to MAP receive second-line treatments such as etoposide and, in more recent years, the kinase inhibitor regorafenib. BCL-2 and its close relatives enforce cellular survival and have been implicated in the development and progression of various cancer types. BH3-mimetics antagonize pro-survival members of the BCL-2 family to directly stimulate apoptosis. These drugs have been proven to be efficacious in other cancer types, but their use in osteosarcoma has been relatively unexplored to date. We investigated the potential efficacy of BH3-mimetics against osteosarcoma cells in vitro and examined their cooperation with regorafenib in vivo. We demonstrated that osteosarcoma cell lines could be killed through inhibition of MCL-1 combined with BCL-2 or BCL-xL antagonism. Inhibition of MCL-1 also sensitized osteosarcoma cells to killing by second-line osteosarcoma treatments, particularly regorafenib. Importantly, we found that inhibition of MCL-1 with the BH3-mimetic S63845 combined with regorafenib significantly prolonged the survival of mice bearing pulmonary osteosarcoma metastases. Together, our results highlight the importance of MCL-1 in osteosarcoma cell survival and present a potential therapeutic avenue that may improve metastatic osteosarcoma patient outcomes.
Collapse
Affiliation(s)
- Yanhao Ji
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Michael A Harris
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Lucas M Newton
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
- Swinburne University, Hawthorn, VIC, 3122, Australia
| | - Tiffany J Harris
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - W Douglas Fairlie
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Erinna F Lee
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Christine J Hawkins
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
5
|
Tambe M, Unterberger S, Kriegbaum MC, Vänttinen I, Olgac EJ, Vähä-Koskela M, Kontro M, Wennerberg K, Heckman CA. Venetoclax triggers sublethal apoptotic signaling in venetoclax-resistant acute myeloid leukemia cells and induces vulnerability to PARP inhibition and azacitidine. Cell Death Dis 2024; 15:750. [PMID: 39414773 PMCID: PMC11484809 DOI: 10.1038/s41419-024-07140-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
Venetoclax plus azacitidine treatment is clinically beneficial for elderly and unfit acute myeloid leukemia (AML) patients. However, the treatment is rarely curative, and relapse due to resistant disease eventually emerges. Since no current clinically feasible treatments are known to be effective at the state of acquired venetoclax resistance, this is becoming a major challenge in AML treatment. Studying venetoclax-resistant AML cell lines, we observed that venetoclax induced sublethal apoptotic signaling and DNA damage even though cell survival and growth were unaffected. This effect could be due to venetoclax inducing a sublethal degree of mitochondrial outer membrane permeabilization. Based on these results, we hypothesized that the sublethal apoptotic signaling induced by venetoclax could constitute a vulnerability in venetoclax-resistant AML cells. This was supported by screens with a broad collection of drugs, where we observed a synergistic effect between venetoclax and PARP inhibition in venetoclax-resistant cells. Additionally, the venetoclax-PARP inhibitor combination prevented the acquisition of venetoclax resistance in treatment naïve AML cell lines. Furthermore, the addition of azacitidine to the venetoclax-PARP inhibitor combination enhanced venetoclax induced DNA damage and exhibited exceptional sensitivity and long-term responses in the venetoclax-resistant AML cell lines and samples from AML patients that had clinically relapsed under venetoclax-azacitidine therapy. In conclusion, we mechanistically identify a new vulnerability in acquired venetoclax-resistant AML cells and identify PARP inhibition as a potential therapeutic approach to overcome acquired venetoclax resistance in AML.
Collapse
MESH Headings
- Humans
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Azacitidine/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Cell Line, Tumor
- DNA Damage/drug effects
- Drug Resistance, Neoplasm/drug effects
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Signal Transduction/drug effects
- Sulfonamides/pharmacology
Collapse
Affiliation(s)
- Mahesh Tambe
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Sarah Unterberger
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Mette C Kriegbaum
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Ida Vänttinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Ezgi June Olgac
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Mika Kontro
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Krister Wennerberg
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
La Marca JE, Kelly GL, Strasser A, Diepstraten ST. Don't fear the reaper: The role of regulated cell death in tumorigenesis and BH3-mimetics for cancer therapy. Dev Cell 2024; 59:2532-2548. [PMID: 39378839 DOI: 10.1016/j.devcel.2024.06.018] [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: 03/01/2024] [Revised: 05/20/2024] [Accepted: 06/20/2024] [Indexed: 10/10/2024]
Abstract
From its earliest characterization, it has been recognized that there is a role for regulated (programmed) cell death in cancer. As our understanding of the different types of programmed cell death processes and their molecular control has advanced, so have the technologies that allow us to manipulate these processes to, for example, fight against cancer. In this review, we describe the roles of the different forms of regulated cell death in the development of cancer as well as their potential therapeutic exploitation. In that vein, we explore the development and use of BH3-mimetics, a unique class of drugs that can directly activate the apoptotic cell death machinery to treat cancer. Finally, we address key challenges that face the field to improve the use of these therapeutics and the efforts that are being undertaken to do so.
Collapse
Affiliation(s)
- John E La Marca
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Genome Engineering and Cancer Modelling Program, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia.
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Andreas Strasser
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Sarah T Diepstraten
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
7
|
Rahman S, Affleck AG, Ruhl RA, Patel RK, Gao L, Brinkerhoff BT, Tsikitis VL, Anand S. Combinatorial Inhibition of Complement Factor D and BCL2 for Early-Onset Colorectal Cancer. Dis Colon Rectum 2024; 67:940-950. [PMID: 38479005 DOI: 10.1097/dcr.0000000000003199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
BACKGROUND The tumor immune microenvironment is distinct between early-onset and late-onset colorectal cancer, which facilitates tumor progression. We previously identified several genes, including complement factor D, as having increased expression in patients with early-onset colorectal cancer. OBJECTIVE This study aimed to assess and validate the differential expression of immune genes in early-onset and late-onset colorectal cancer. We also aimed to test known drugs targeting genes increased in early-onset colorectal cancer in preclinical mouse models. DESIGN A retrospective cohort study with analysis was performed using tumor RNA from formalin-fixed paraffin-embedded cell culture and immunohistochemistry to validate gene expression and function and in vivo preclinical tumor study to assess drug efficacy. SETTINGS The Oregon Colorectal Cancer Registry was queried to identify patients with colorectal cancer. PATIENTS The study included 67 patients with early-onset colorectal cancer and 54 patients with late-onset colorectal cancer. INTERVENTIONS Preclinical animal models using the HCT-116 colon cancer cell line were treated with the complement factor D inhibitor danicopan and the BCL2 inhibitor venetoclax, or with vehicle controls. MAIN OUTCOME MEASURES Elevated RNA signatures using NanoString data were evaluated by the retrospective cohort. When inhibiting these markers in the mouse preclinical model, tumor volume and weight were the main outcome measures. RESULTS After updating our sample size from our previously published data, we found that complement factor D and BCL2, genes with known function and small molecule inhibitors, are elevated in patients with early-onset colorectal cancer. When inhibiting these markers with the drugs danicopan and venetoclax in a mouse model, we found that the combination of these drugs decreased tumor burden but also resulted in toxicity. LIMITATIONS This study is limited by a small sample size and a subcutaneous tumor model. CONCLUSIONS Combinatorial inhibition of early-onset associated genes complement factor D and BCL2 slows the growth of early-onset colorectal cancer in a mouse preclinical model. See Video Abstract . INHIBICIN COMBINADA DEL FACTOR DCOMPLEMENTARIO Y DEL BCL EN CASOS DE CNCER COLORRECTAL DE APARICIN TEMPRANA ANTECEDENTES:El microambiente inmunológico del tumor es distinto entre el cáncer colorrectal de aparición temprana y el de aparición tardía, lo que facilita la progresión de dicho tumor. Anteriormente identificamos varios genes, incluidos el factor D-Complementario, con una mayor expresión en pacientes con cáncer colorrectal de aparición temprana.OBJETIVO:El presente estudio tuvo como objetivo el evaluar y validar la expresión diferenciada de genes inmunes en casos de cáncer colorrectal de aparición temprana y tardía. También nos propusimos evaluar los fármacos conocidos dirigidos sobre los genes aumentados en el cáncer colorrectal de aparición temprana en modelos pre-clínicos en ratones.DISEÑO:Estudio de cohortes con análisis retrospectivo utilizando el ARN tumoral procedente de cultivos celulares fijados con formalina e incluidos en parafina, y el analisis por inmunohistoquímica para validar la expresión y la función genética. Se realizó el estudio pre-clínico de los tumores in vivo para evaluar la eficacia de los fármacos.AJUSTES:Se consultó el Registro de Oregon de casos de Cáncer Colorrectal para encontrar los pacientes afectados.SUJETOS:67 pacientes con cáncer colorrectal de aparición temprana y 54 pacientes con cáncer colorrectal de aparición tardía.INTERVENCIONES (SI LAS HUBIESE):Los modelos animales pre-clínicos que utilizaron la línea celular de cáncer de colon HCT-116 se trataron con el inhibidor del factor D-Complementario o Danicopan y con el inhibidor de BCL-2 o Venetoclax, ambos con control del transportador.PRINCIPALES MEDIDAS DE RESULTADO:Se evaluaron las firmas de ARN elevadas utilizando los datos del NanoString a partir de la cohorte retrospectiva. Al inhibir estos marcadores del modelo pre-clínico en los ratones, el volumen y el peso del tumor fueron las principales medidas de resultado.RESULTADOS:Después de actualizar el tamaño de nuestra muestra a partir de datos publicados con anterioridad, encontramos que el factor D-Complementario y BCL-2, genes con función conocida e inhibidores de moléculas pequeñas, se encuentran elevados en aquellos pacientes con cáncer colorrectal de aparición temprana. Al inhibir estos marcadores con los medicamentos Danicopan y Venetoclax en el modelo de ratones vivos, encontramos que la combinación de estos dos farmacos disminuyó la carga tumoral pero también produjo toxicidad.LIMITACIONES:Estudio limitado por un tamaño de muestra pequeño y el modelo de tumor subcutáneo.CONCLUSIONES:La inhibición combinada de genes asociados de aparición temprana, el factor D-Complementario y el BCL-2, enlentecen el crecimiento del cáncer colorrectal de aparición temprana del modelo preclínico en ratones. (Traducción-Dr. Xavier Delgadillo ).
Collapse
Affiliation(s)
- Shahrose Rahman
- Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Arthur G Affleck
- Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Rebecca A Ruhl
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Ranish K Patel
- Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Lina Gao
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Brian T Brinkerhoff
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, Oregon
| | | | - Sudarshan Anand
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Department of Radiation Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
8
|
Fitzgerald MC, O'Halloran PJ, Kerrane SA, Ní Chonghaile T, Connolly NMC, Murphy BM. The identification of BCL-XL and MCL-1 as key anti-apoptotic proteins in medulloblastoma that mediate distinct roles in chemotherapy resistance. Cell Death Dis 2023; 14:705. [PMID: 37898609 PMCID: PMC10613306 DOI: 10.1038/s41419-023-06231-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 09/25/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Medulloblastoma is the most common malignant paediatric brain tumour, representing 20% of all paediatric intercranial tumours. Current aggressive treatment protocols and the use of radiation therapy in particular are associated with high levels of toxicity and significant adverse effects, and long-term sequelae can be severe. Therefore, improving chemotherapy efficacy could reduce the current reliance on radiation therapy. Here, we demonstrated that systems-level analysis of basal apoptosis protein expression and their signalling interactions can differentiate between medulloblastoma cell lines that undergo apoptosis in response to chemotherapy, and those that do not. Combining computational predictions with experimental BH3 profiling, we identified a therapeutically-exploitable dependence of medulloblastoma cells on BCL-XL, and experimentally validated that BCL-XL targeting, and not targeting of BCL-2 or MCL-1, can potentiate cisplatin-induced cytotoxicity in medulloblastoma cell lines with low sensitivity to cisplatin treatment. Finally, we identified MCL-1 as an anti-apoptotic mediator whose targeting is required for BCL-XL inhibitor-induced apoptosis. Collectively, our study identifies that BCL-XL and MCL-1 are the key anti-apoptotic proteins in medulloblastoma, which mediate distinct protective roles. While BCL-XL has a first-line role in protecting cells from apoptosis basally, MCL-1 represents a second line of defence that compensates for BCL-XL upon its inhibition. We provide rationale for the further evaluation of BCL-XL and MCL-1 inhibitors in the treatment of medulloblastoma, and together with current efforts to improve the cancer-specificity of BCL-2 family inhibitors, these novel treatment strategies have the potential to improve the future clinical management of medulloblastoma.
Collapse
Affiliation(s)
- Marie-Claire Fitzgerald
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- National Children's Research Centre at the Children's Health Ireland at Crumlin, Dublin, D12 N512, Ireland
| | - Philip J O'Halloran
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham, UK
| | - Sean A Kerrane
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- National Children's Research Centre at the Children's Health Ireland at Crumlin, Dublin, D12 N512, Ireland
| | - Triona Ní Chonghaile
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
| | - Niamh M C Connolly
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
| | - Brona M Murphy
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland.
- National Children's Research Centre at the Children's Health Ireland at Crumlin, Dublin, D12 N512, Ireland.
| |
Collapse
|
9
|
Diepstraten ST, La Marca JE, Chang C, Young S, Strasser A, Kelly GL. BCL-W makes only minor contributions to MYC-driven lymphoma development. Oncogene 2023; 42:2776-2781. [PMID: 37567974 PMCID: PMC10491490 DOI: 10.1038/s41388-023-02804-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/25/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023]
Abstract
The BH3-mimetic drug Venetoclax, a specific inhibitor of anti-apoptotic BCL-2, has had clinical success for the treatment of chronic lymphocytic leukaemia and acute myeloid leukaemia. Attention has now shifted towards related pro-survival BCL-2 family members, hypothesising that new BH3-mimetic drugs targeting these proteins may emulate the success of Venetoclax. BH3-mimetics targeting pro-survival MCL-1 or BCL-XL have entered clinical trials, but managing on-target toxicities is challenging. While increasing evidence suggests BFL-1/A1 is a resistance factor for diverse chemotherapeutic agents and BH3-mimetic drugs in haematological malignancies, few studies have explored the role of BCL-W in the development, expansion, and therapeutic responses of cancer. Previously, we found that BCL-W was not required for the ongoing survival and growth of various established human Burkitt lymphoma and diffuse large B cell lymphoma cell lines. However, questions remained about whether BCL-W impacts lymphoma development. Here, we show that BCL-W appears dispensable for MYC-driven lymphomagenesis, and such tumours arising in the absence of BCL-W show no compensatory changes to BCL-2 family member expression, nor altered sensitivity to BH3-mimetic drugs. These results demonstrate that BCL-W does not play a major role in the development of MYC-driven lymphoma or the responses of these tumours to anti-cancer agents.
Collapse
Affiliation(s)
- Sarah T Diepstraten
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - John E La Marca
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Catherine Chang
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
| | - Savannah Young
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
| | - Andreas Strasser
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Gemma L Kelly
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
10
|
Chiou JT, Wu YY, Lee YC, Chang LS. BCL2L1 inhibitor A-1331852 inhibits MCL1 transcription and triggers apoptosis in acute myeloid leukemia cells. Biochem Pharmacol 2023; 215:115738. [PMID: 37562509 DOI: 10.1016/j.bcp.2023.115738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
BH3 mimetics exert anticancer activity by inhibiting anti-apoptotic BCL2 proteins. However, accumulating evidence indicates that the off-target effects of these drugs tightly modulates their anticancer activities. In this study, we investigated whether the BCL2L1 inhibitor A-1331852 induced the death of U937 acute myeloid leukemia (AML) cells through a non-BCL2L1-targeted effect. A-1331852-induced apoptosis in U937 cells was characterized by increased ROS production, downregulation of MCL1, and loss of mitochondrial membrane potential. Ectopic expression of MCL1 alleviated A-1331852-induced mitochondrial depolarization and cytotoxicity in U937 cells. A-1331852-induced ROS production increased p38 MAPK phosphorylation and inhibited MCL1 transcription. Inhibition of p38 MAPK activation restored MCL1 expression in A-1331852-treated cells. A-1331852 triggered p38 MAPK-mediated Cullin 3 downregulation, which in turn increased PP2Acα expression, thereby reducing CREB phosphorylation. A-1331852 reduced the binding of CREB to the MCL1 promoter, leading to the inhibition of CREB-mediated MCL1 transcription. Furthermore, A-1331852 acted synergistically with the BCL2 inhibitor ABT-199 to induce U937 and ABT-199-resistant U937 cell death by inhibiting MCL1 expression. A similar phenomenon caused A-1331852-induced MCL1 downregulation and cytotoxicity in AML HL-60 cells. Collectively, our data suggest that A-1331852 shows an off-target effect of inhibiting MCL1 transcription, ultimately leading to U937 and HL-60 cell death.
Collapse
Affiliation(s)
- Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yu-Ying Wu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
11
|
Michalski M, Bauer M, Walz F, Tümen D, Heumann P, Stöckert P, Gunckel M, Kunst C, Kandulski A, Schmid S, Müller M, Gülow K. Simultaneous Inhibition of Mcl-1 and Bcl-2 Induces Synergistic Cell Death in Hepatocellular Carcinoma. Biomedicines 2023; 11:1666. [PMID: 37371761 PMCID: PMC10295989 DOI: 10.3390/biomedicines11061666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Despite the recent approval of new therapies, the prognosis for patients with hepatocellular carcinoma (HCC) remains poor. There is a clinical need for new highly effective therapeutic options. Here, we present a combined application of BH3-mimetics as a potential new treatment option for HCC. BH3-mimetics inhibit anti-apoptotic proteins of the BCL-2 family and, thus, trigger the intrinsic apoptosis pathway. Anti-apoptotic BCL-2 proteins such as Bcl-2 and Mcl-1 are frequently overexpressed in HCC. Therefore, we analyzed the efficacy of the two BH3-mimetics ABT-199 (Bcl-2 inhibitor) and MIK665 (Mcl-1 inhibitor) in HCC cell lines with differential expression levels of endogenous Bcl-2 and Mcl-1. While administration of one BH3-mimetic alone did not substantially trigger cell death, the combination of two inhibitors enhanced induction of the intrinsic apoptosis pathway. Both drugs acted synergistically, highlighting the effectivity of this specific BH3-mimetic combination, particularly in HCC cell lines. These results indicate the potential of combining inhibitors of the BCL-2 family as new therapeutic options in HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Karsten Gülow
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; (M.M.); (M.B.); (F.W.); (D.T.); (P.H.); (P.S.); (M.G.); (C.K.); (A.K.); (S.S.); (M.M.)
| |
Collapse
|
12
|
Wei Y, Zhang L, Wang C, Li Z, Luo M, Xie G, Yang X, Li M, Ren S, Zhao D, Gao R, Gong J. Anti-apoptotic protein BCL-XL as a therapeutic vulnerability in gastric cancer. Animal Model Exp Med 2023; 6:245-254. [PMID: 37271936 PMCID: PMC10272913 DOI: 10.1002/ame2.12330] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND New therapeutic targets are needed to improve the outcomes for gastric cancer (GC) patients with advanced disease. Evasion of programmed cell death (apoptosis) is a hallmark of cancer cells and direct induction of apoptosis by targeting the pro-survival BCL2 family proteins represents a promising therapeutic strategy for cancer treatment. Therefore, understanding the molecular mechanisms underpinning cancer cell survival could provide a molecular basis for potential therapeutic interventions. METHOD Here we explored the role of BCL2L1 and the encoded anti-apoptotic BCL-XL in GC. Using Droplet Digital PCR (ddPCR) technology to investigate the DNA amplification of BCL2L1 in GC samples and GC cell lines, the sensitivity of GC cell lines to selective BCL-XL inhibitors A1155463 and A1331852, pan-inhibitor ABT-263, and VHL-based PROTAC-BCL-XL was analyzed using (CellTiter-Glo) CTG assay in vitro. Western Blot (WB) was used to detect the protein expression of BCL2 family members in GC cell lines and the manner in which PROTAC-BCL-XL kills GC cells. Co-immunoprecipitation (Co-IP) was used to investigate the mechanism of A1331852 and ABT-263 kills GC cell lines. DDPCR, WB, and real-time PCR (RTPCR) were used to investigate the correlation between DNA, RNA, protein levels, and drug activity. RESULTS The functional assay showed that a subset of GC cell lines relies on BCL-XL for survival. In gastric cancer cell lines, BCL-XL inhibitors A1155463 and A1331852 are more sensitive than the pan BCL2 family inhibitor ABT-263, indicating that ABT-263 is not an optimal inhibitor of BCL-XL. VHL-based PROTAC-BCL-XL DT2216 appears to be active in GC cells. DT2216 induces apoptosis of gastric cancer cells in a time- and dose-dependent manner through the proteasome pathway. Statistical analysis showed that the BCL-XL protein level predicts the response of GC cells to BCL-XL targeting therapy and BCL2L1 gene CNVs do not reliably predict BCL-XL expression. CONCLUSION We identified BCL-XL as a promising therapeutic target in a subset of GC cases with high levels of BCL-XL protein expression. Functionally, we demonstrated that both selective BCL-XL inhibitors and VHL-based PROTAC BCL-XL can potently kill GC cells that are reliant on BCL-XL for survival. However, we found that BCL2L1 copy number variations (CNVs) cannot reliably predict BCL-XL expression, but the BCL-XL protein level serves as a useful biomarker for predicting the sensitivity of GC cells to BCL-XL-targeting compounds. Taken together, our study pinpointed BCL-XL as potential druggable target for specific subsets of GC.
Collapse
Affiliation(s)
- Yumin Wei
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Liping Zhang
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Chao Wang
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Zefeng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mingjie Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Guomin Xie
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Xingjiu Yang
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Mengyuan Li
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Shuyue Ren
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Dongbing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ran Gao
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Jia‐Nan Gong
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| |
Collapse
|
13
|
Selt F, Sigaud R, Valinciute G, Sievers P, Zaman J, Alcon C, Schmid S, Peterziel H, Tsai JW, Guiho R, Martínez-Barbera JP, Pusch S, Deng J, Zhai Y, van Tilburg CM, Schuhman MU, El Damaty A, Bandopadhayay P, Herold-Mende C, von Deimling A, Pfister SM, Montero J, Capper D, Oehme I, Sahm F, Jones DTW, Witt O, Milde T. BH3 mimetics targeting BCL-XL impact the senescent compartment of pilocytic astrocytoma. Neuro Oncol 2023; 25:735-747. [PMID: 35977048 PMCID: PMC10076946 DOI: 10.1093/neuonc/noac199] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pilocytic astrocytoma (PA) is the most common pediatric brain tumor and a mitogen-activated protein kinase (MAPK)-driven disease. Oncogenic MAPK-signaling drives the majority of cells into oncogene-induced senescence (OIS). While OIS induces resistance to antiproliferative therapies, it represents a potential vulnerability exploitable by senolytic agents. METHODS We established new patient-derived PA cell lines that preserve molecular features of the primary tumors and can be studied in OIS and proliferation depending on expression or repression of the SV40 large T antigen. We determined expression of anti-apoptotic BCL-2 members in these models and primary PA. Dependence of senescent PA cells on anti-apoptotic BCL-2 members was investigated using a comprehensive set of BH3 mimetics. RESULTS Senescent PA cells upregulate BCL-XL upon senescence induction and show dependency on BCL-XL for survival. BH3 mimetics with high affinity for BCL-XL (BCL-XLi) reduce metabolic activity and induce mitochondrial apoptosis in senescent PA cells at nano-molar concentrations. In contrast, BH3 mimetics without BCL-XLi activity, conventional chemotherapy, and MEK inhibitors show no effect. CONCLUSIONS Our data demonstrate that BCL-XL is critical for survival of senescent PA tumor cells and provides proof-of-principle for the use of clinically available BCL-XL-dependent senolytics.
Collapse
Affiliation(s)
- Florian Selt
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Romain Sigaud
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Gintvile Valinciute
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Philipp Sievers
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Zaman
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clara Alcon
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Simone Schmid
- Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Berlin, Berlin, Germany
| | - Heike Peterziel
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Jessica W Tsai
- Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Boston, Massachusetts, USA
| | - Romain Guiho
- Developmental Biology and Cancer Research & Teaching Department, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Juan Pedro Martínez-Barbera
- Developmental Biology and Cancer Research & Teaching Department, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Stefan Pusch
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jing Deng
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, Jiangsu Province, China
| | - Yifan Zhai
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, Jiangsu Province, China
| | - Cornelis M van Tilburg
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin U Schuhman
- Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Ahmed El Damaty
- Pediatric Neurosurgery Division, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Pratiti Bandopadhayay
- Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Joan Montero
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - David Capper
- Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Berlin, Berlin, Germany
| | - Ina Oehme
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Felix Sahm
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David T W Jones
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Till Milde
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
14
|
Kong S, Moharil P, Handly‐Santana A, Boehnke N, Panayiotou R, Gomerdinger V, Covarrubias G, Pires IS, Zervantonakis I, Brugge J, Hammond PT. Synergistic combination therapy delivered via layer-by-layer nanoparticles induces solid tumor regression of ovarian cancer. Bioeng Transl Med 2023; 8:e10429. [PMID: 36925689 PMCID: PMC10013771 DOI: 10.1002/btm2.10429] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/11/2022] Open
Abstract
The majority of patients with high grade serous ovarian cancer (HGSOC) develop recurrent disease and chemotherapy resistance. To identify drug combinations that would be effective in treatment of chemotherapy resistant disease, we examined the efficacy of drug combinations that target the three antiapoptotic proteins most commonly expressed in HGSOC-BCL2, BCL-XL, and MCL1. Co-inhibition of BCL2 and BCL-XL (ABT-263) with inhibition of MCL1 (S63845) induces potent synergistic cytotoxicity in multiple HGSOC models. Since this drug combination is predicted to be toxic to patients due to the known clinical morbidities of each drug, we developed layer-by-layer nanoparticles (LbL NPs) that co-encapsulate these inhibitors in order to target HGSOC tumor cells and reduce systemic toxicities. We show that the LbL NPs can be designed to have high association with specific ovarian tumor cell types targeted in these studies, thus enabling a more selective uptake when delivered via intraperitoneal injection. Treatment with these LbL NPs displayed better potency than free drugs in vitro and resulted in near-complete elimination of solid tumor metastases of ovarian cancer xenografts. Thus, these results support the exploration of LbL NPs as a strategy to deliver potent drug combinations to recurrent HGSOC. While these findings are described for co-encapsulation of a BCL2/XL and a MCL1 inhibitor, the modular nature of LbL assembly provides flexibility in the range of therapies that can be incorporated, making LbL NPs an adaptable vehicle for delivery of additional combinations of pathway inhibitors and other oncology drugs.
Collapse
Affiliation(s)
- Stephanie Kong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeMassachusettsUnited States
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUnited States
| | - Pearl Moharil
- Harvard Medical SchoolHarvard UniversityBostonMassachusettsUnited States
| | | | - Natalie Boehnke
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeMassachusettsUnited States
| | - Richard Panayiotou
- Harvard Medical SchoolHarvard UniversityBostonMassachusettsUnited States
| | - Victoria Gomerdinger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeMassachusettsUnited States
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUnited States
| | - Gil Covarrubias
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeMassachusettsUnited States
| | - Ivan S. Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeMassachusettsUnited States
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUnited States
| | - Ioannis Zervantonakis
- Harvard Medical SchoolHarvard UniversityBostonMassachusettsUnited States
- Department of BioengineeringUniversity of PittsburghPittsburghPennsylvaniaUnited States
| | - Joan Brugge
- Harvard Medical SchoolHarvard UniversityBostonMassachusettsUnited States
| | - Paula T. Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeMassachusettsUnited States
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUnited States
| |
Collapse
|
15
|
Diepstraten ST, Young S, La Marca JE, Wang Z, Kluck RM, Strasser A, Kelly GL. Lymphoma cells lacking pro-apoptotic BAX are highly resistant to BH3-mimetics targeting pro-survival MCL-1 but retain sensitivity to conventional DNA-damaging drugs. Cell Death Differ 2023; 30:1005-1017. [PMID: 36755070 PMCID: PMC10070326 DOI: 10.1038/s41418-023-01117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
BH3-mimetic drugs are an anti-cancer therapy that can induce apoptosis in malignant cells by directly binding and inhibiting pro-survival proteins of the BCL-2 family. The BH3-mimetic drug venetoclax, which targets BCL-2, has been approved for the treatment of chronic lymphocytic leukaemia and acute myeloid leukaemia by regulatory authorities worldwide. However, while most patients initially respond well, resistance and relapse while on this drug is an emerging and critical issue in the clinic. Though some studies have begun uncovering the factors involved in resistance to BCL-2-targeting BH3-mimetic drugs, little focus has been applied to pre-emptively tackle resistance for the next generation of BH3-mimetic drugs targeting MCL-1, which are now in clinical trials for diverse blood cancers. Therefore, using pre-clinical mouse and human models of aggressive lymphoma, we sought to predict factors likely to contribute to the development of resistance in patients receiving MCL-1-targeting BH3-mimetic drugs. First, we performed multiple whole genome CRISPR/Cas9 KO screens and identified that loss of the pro-apoptotic effector protein BAX, but not its close relative BAK, could confer resistance to MCL-1-targeting BH3-mimetic drugs in both short-term and long-term treatment regimens, even in lymphoma cells lacking the tumour suppressor TRP53. Furthermore, we found that mouse Eµ-Myc lymphoma cells selected for loss of BAX, as well as upregulation of the untargeted pro-survival BCL-2 family proteins BCL-XL and A1, when made naturally resistant to MCL-1 inhibitors by culturing them in increasing doses of drug over time, a situation mimicking the clinical application of these drugs. Finally, we identified therapeutic approaches which could overcome these two methods of resistance: the use of chemotherapeutic drugs or combined BH3-mimetic treatment, respectively. Collectively, these results uncover some key factors likely to cause resistance to MCL-1 inhibition in the clinic and suggest rational therapeutic strategies to overcome resistance that should be investigated further.
Collapse
Affiliation(s)
- Sarah T Diepstraten
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Savannah Young
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - John E La Marca
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Zilu Wang
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Ruth M Kluck
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
16
|
Abstract
The intrinsic apoptosis pathway is controlled by the BCL-2 family of proteins. Although the pro-survival members of this family can help cancer cells evade apoptosis, they may also produce apoptotic vulnerabilities that can potentially be exploited therapeutically. Apoptotic vulnerabilities can be driven by endogenous factors including altered genetics, signaling, metabolism, structure and lineage or differentiation state as well as imposed factors, the most prominent being exposure to anti-cancer agents. The recent development of BH3 mimetics that inhibit pro-survival BCL-2 family proteins has allowed these apoptotic vulnerabilities to be targeted with demonstrable clinical success. Here, we review the key concepts that are vital for understanding, uncovering, and exploiting apoptotic vulnerabilities in cancer for the potential improvement of patient outcomes.
Collapse
Affiliation(s)
- Kristopher A. Sarosiek
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kris C. Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| |
Collapse
|
17
|
Khan S, Kellish P, Connis N, Thummuri D, Wiegand J, Zhang P, Zhang X, Budamagunta V, Hua N, Yang Y, De U, Jin L, Zhang W, Zheng G, Hromas R, Hann C, Zajac-Kaye M, Kaye FJ, Zhou D. Co-targeting BCL-X L and MCL-1 with DT2216 and AZD8055 synergistically inhibit small-cell lung cancer growth without causing on-target toxicities in mice. Cell Death Dis 2023; 9:1. [PMID: 36588105 PMCID: PMC9806104 DOI: 10.1038/s41420-022-01296-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023]
Abstract
Small-cell lung cancer (SCLC) is an aggressive malignancy with limited therapeutic options. The dismal prognosis in SCLC is in part associated with an upregulation of BCL-2 family anti-apoptotic proteins, including BCL-XL and MCL-1. Unfortunately, the currently available inhibitors of BCL-2 family anti-apoptotic proteins, except BCL-2 inhibitors, are not clinically relevant because of various on-target toxicities. We, therefore, aimed to develop an effective and safe strategy targeting these anti-apoptotic proteins with DT2216 (our platelet-sparing BCL-XL degrader) and AZD8055 (an mTOR inhibitor) to avoid associated on-target toxicities while synergistically optimizing tumor response. Through BH3 mimetic screening, we identified a subset of SCLC cell lines that is co-dependent on BCL-XL and MCL-1. After screening inhibitors of selected tumorigenic pathways, we found that AZD8055 selectively downregulates MCL-1 in SCLC cells and its combination with DT2216 synergistically killed BCL-XL/MCL-1 co-dependent SCLC cells, but not normal cells. Mechanistically, the combination caused BCL-XL degradation and suppression of MCL-1 expression, and thus disrupted MCL-1 interaction with BIM leading to an enhanced apoptotic induction. In vivo, the DT2216 + AZD8055 combination significantly inhibited the growth of cell line-derived and patient-derived xenografts and reduced tumor burden accompanied by increased survival in a genetically engineered mouse model of SCLC without causing appreciable thrombocytopenia or other normal tissue injuries. Thus, these preclinical findings lay a strong foundation for future clinical studies to test DT2216 + mTOR inhibitor combinations in a subset of SCLC patients whose tumors are co-driven by BCL-XL and MCL-1.
Collapse
Affiliation(s)
- Sajid Khan
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA. .,Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA. .,Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Patrick Kellish
- grid.15276.370000 0004 1936 8091Department of Anatomy & Cell Biology, College of Medicine, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Nick Connis
- grid.21107.350000 0001 2171 9311Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD USA
| | - Dinesh Thummuri
- grid.15276.370000 0004 1936 8091Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Janet Wiegand
- grid.15276.370000 0004 1936 8091Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Peiyi Zhang
- grid.15276.370000 0004 1936 8091Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Xuan Zhang
- grid.15276.370000 0004 1936 8091Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Vivekananda Budamagunta
- grid.15276.370000 0004 1936 8091Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL USA
| | - Nan Hua
- grid.15276.370000 0004 1936 8091Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Yang Yang
- grid.267309.90000 0001 0629 5880Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX USA ,grid.15276.370000 0004 1936 8091Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Umasankar De
- grid.15276.370000 0004 1936 8091Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| | - Lingtao Jin
- grid.267309.90000 0001 0629 5880Department of Molecular Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX USA
| | - Weizhou Zhang
- grid.15276.370000 0004 1936 8091Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| | - Guangrong Zheng
- grid.15276.370000 0004 1936 8091Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Robert Hromas
- grid.267309.90000 0001 0629 5880Department of Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX USA
| | - Christine Hann
- grid.21107.350000 0001 2171 9311Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD USA
| | - Maria Zajac-Kaye
- grid.15276.370000 0004 1936 8091Department of Anatomy & Cell Biology, College of Medicine, University of Florida, Gainesville, FL USA
| | - Frederic J. Kaye
- grid.15276.370000 0004 1936 8091Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| | - Daohong Zhou
- grid.267309.90000 0001 0629 5880Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX USA ,grid.267309.90000 0001 0629 5880Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX USA ,grid.15276.370000 0004 1936 8091Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL USA
| |
Collapse
|
18
|
Wang N, Hakimi AA, Chen Y. XL-ing at Induction of Apoptosis in Kidney Cancer through Inhibition of BCL-XL. Clin Cancer Res 2022; 28:4600-4602. [PMID: 35984355 PMCID: PMC9633424 DOI: 10.1158/1078-0432.ccr-22-2104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/02/2022] [Accepted: 08/09/2022] [Indexed: 01/24/2023]
Abstract
Through analysis of the cancer dependency map of CRISPR and short hairpin RNA datasets, the antiapoptotic BCL-XL was found to be a selective dependency in kidney cancer. Among kidney cancers, BCL-XL inhibition is most active in those with a mesenchymal gene signature, which portends a poor prognosis and response to current therapies. See related article by Grubb et al., p. 4689.
Collapse
Affiliation(s)
- Naitao Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - A. Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York NY 10065, USA,Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA,Department of Medicine, Genitourinary Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA,Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
19
|
Grubb T, Maganti S, Krill-Burger JM, Fraser C, Stransky L, Radivoyevitch T, Sarosiek KA, Vazquez F, Kaelin WG, Chakraborty AA. A Mesenchymal Tumor Cell State Confers Increased Dependency on the BCL-XL Antiapoptotic Protein in Kidney Cancer. Clin Cancer Res 2022; 28:4689-4701. [PMID: 35776130 PMCID: PMC9633392 DOI: 10.1158/1078-0432.ccr-22-0669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/07/2022] [Accepted: 06/28/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Advanced/metastatic forms of clear-cell renal cell carcinomas (ccRCC) have limited therapeutic options. Genome-wide genetic screens have identified cellular dependencies in many cancers. Using the Broad Institute/Novartis combined short hairpin RNA (shRNA) dataset, and cross-validation with the CRISPR/Cas9 DepMap (21Q3) dataset, we sought therapeutically actionable dependencies in kidney lineage cancers. EXPERIMENTAL DESIGN We identified preferential genetic dependencies in kidney cancer cells versus other lineages. BCL2L1, which encodes the BCL-XL antiapoptotic protein, scored as the top actionable dependency. We validated this finding using genetic and pharmacologic tools in a panel of ccRCC cell lines. Select BCL-XL-dependent (versus independent) cell lines were then transcriptionally profiled to identify biomarkers and mechanistic drivers of BCL-XL dependence. Cell-based studies (in vitro and in vivo) and clinical validations were used to address physiologic relevance. RESULTS Inactivation of BCL-XL, but not BCL-2, led to fitness defects in renal cancer cells, and sensitized them to chemotherapeutics. Transcriptomic profiling identified a "BCL-XL dependency" signature, including an elevated mesenchymal gene signature. A mesenchymal state was both necessary and sufficient to confer increased BCL-XL dependence. The "BCL-XL dependency" signature was observed in approximately 30% of human ccRCCs, which were also associated with worse clinical outcomes. Finally, an orally bioavailable BCL-XL inhibitor, A-1331852, showed antitumor efficacy in vivo. CONCLUSIONS Our studies uncovered an unexpected link between cell state and BCL-XL dependence in ccRCC. Therapeutic agents that specifically target BCL-XL are available. Our work justifies testing the utility of BCL-XL blockade to target, likely, a clinically aggressive subset of human kidney cancers. See related commentary by Wang et al., p. 4600.
Collapse
Affiliation(s)
- Treg Grubb
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Smruthi Maganti
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Cameron Fraser
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.,Molecular and Integrative Physiology Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Laura Stransky
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Tomas Radivoyevitch
- Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kristopher A. Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.,Molecular and Integrative Physiology Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,Corresponding Authors: William G. Kaelin, Jr., Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA 02115. . Phone: 617-632-3975; Abhishek A. Chakraborty, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195. . Phone: 216-445-6620
| | - Abhishek A. Chakraborty
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Corresponding Authors: William G. Kaelin, Jr., Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA 02115. . Phone: 617-632-3975; Abhishek A. Chakraborty, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195. . Phone: 216-445-6620
| |
Collapse
|
20
|
Rosa N, Speelman-Rooms F, Parys JB, Bultynck G. Modulation of Ca 2+ signaling by antiapoptotic Bcl-2 versus Bcl-xL: From molecular mechanisms to relevance for cancer cell survival. Biochim Biophys Acta Rev Cancer 2022; 1877:188791. [PMID: 36162541 DOI: 10.1016/j.bbcan.2022.188791] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022]
Abstract
Members of the Bcl-2-protein family are key controllers of apoptotic cell death. The family is divided into antiapoptotic (including Bcl-2 itself, Bcl-xL, Mcl-1, etc.) and proapoptotic members (Bax, Bak, Bim, Bim, Puma, Noxa, Bad, etc.). These proteins are well known for their canonical role in the mitochondria, where they control mitochondrial outer membrane permeabilization and subsequent apoptosis. However, several proteins are recognized as modulators of intracellular Ca2+ signals that originate from the endoplasmic reticulum (ER), the major intracellular Ca2+-storage organelle. More than 25 years ago, Bcl-2, the founding member of the family, was reported to control apoptosis through Ca2+ signaling. Further work elucidated that Bcl-2 directly targets and inhibits inositol 1,4,5-trisphosphate receptors (IP3Rs), thereby suppressing proapoptotic Ca2+ signaling. In addition to Bcl-2, Bcl-xL was also shown to impact cell survival by sensitizing IP3R function, thereby promoting prosurvival oscillatory Ca2+ release. However, new work challenges this model and demonstrates that Bcl-2 and Bcl-xL can both function as inhibitors of IP3Rs. This suggests that, depending on the cell context, Bcl-xL could support very distinct Ca2+ patterns. This not only raises several questions but also opens new possibilities for the treatment of Bcl-xL-dependent cancers. In this review, we will discuss the similarities and divergences between Bcl-2 and Bcl-xL regarding Ca2+ homeostasis and IP3R modulation from both a molecular and a functional point of view, with particular emphasis on cancer cell death resistance mechanisms.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Femke Speelman-Rooms
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium.
| |
Collapse
|
21
|
Winter JM, Yadav T, Rutter J. Stressed to death: Mitochondrial stress responses connect respiration and apoptosis in cancer. Mol Cell 2022; 82:3321-3332. [PMID: 35961309 PMCID: PMC9481690 DOI: 10.1016/j.molcel.2022.07.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/27/2022] [Accepted: 07/18/2022] [Indexed: 11/30/2022]
Abstract
Mitochondrial energetics and respiration have emerged as important factors in how cancer cells respond to or evade apoptotic signals. The study of the functional connection between these two processes may provide insight into following questions old and new: how might we target respiration or downstream signaling pathways to amplify apoptotic stress in the context of cancer therapy? Why are respiration and apoptotic regulation housed in the same organelle? Here, we briefly review mitochondrial respiration and apoptosis and then focus on how the intersection of these two processes is regulated by cytoplasmic signaling pathways such as the integrated stress response.
Collapse
Affiliation(s)
- Jacob M Winter
- Department of Biochemistry, Spencer Fox Eccles School of Medicine, The University of Utah, Salt Lake City, UT, USA
| | - Tarun Yadav
- Department of Biochemistry, Spencer Fox Eccles School of Medicine, The University of Utah, Salt Lake City, UT, USA; Department of Biology, Indian Institute of Science Education and Research, Pune, Maharashtra, India
| | - Jared Rutter
- Department of Biochemistry, Spencer Fox Eccles School of Medicine, The University of Utah, Salt Lake City, UT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
22
|
Qian L, Vallega KA, Yao W, Wang D, Zhai Y, He X, Sun SY. Therapeutic potential of the novel Bcl-2/Bcl-X L dual inhibitor, APG1252, alone or in combination against non-small cell lung cancer. Mol Carcinog 2022; 61:1031-1042. [PMID: 36066010 DOI: 10.1002/mc.23458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/08/2022] [Accepted: 08/21/2022] [Indexed: 11/08/2022]
Abstract
Targeting the induction of apoptosis is a promising cancer therapeutic strategy with some clinical success. This study focused on evaluating the therapeutic efficacy of the novel Bcl-2/Bcl-XL dual inhibitor, APG1252-M1 (also named APG-1244; an in vivo active metabolite of APG1252 or pelcitoclax), as a single agent or in combination, against non-small cell lung cancer (NSCLC) cells. APG1252-M1 effectively decreased the survival of some NSCLC cell lines expressing low levels of Mcl-1 and induced apoptosis. Overexpression of ectopic Mcl-1 in the sensitive cells substantially compromised APG1252-M1's cell-killing effects, whereas inhibition of Mcl-1 greatly sensitized insensitive cell lines to APG1252-M1, indicating the critical role of Mcl-1 levels in impacting cell response to APG1252-M1. Moreover, APG1252-M1, when combined with the third generation epidermal growth factor receptor (EGFR) inhibitor, osimertinib, synergistically decreased the survival of EGFR-mutant NSCLC cell lines including those resistant to osimertinib with enhanced induction of apoptosis and abrogated emergence of acquired resistance to osimertinib. Importantly, the combination was effective in inhibiting the growth of osimertinib-resistant tumors in vivo. Collectively, these results demonstrate the efficacy of APG1252 alone or in combination against human NSCLC cells.
Collapse
Affiliation(s)
- Luxi Qian
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China.,Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Karin A Vallega
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Weilong Yao
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA.,Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Dongsheng Wang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Yifan Zhai
- Ascentage Pharma (Suzhou) Co., Ltd, Suzhou, Jiangsu, P. R. China
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| |
Collapse
|
23
|
Ohtsu A, Arai S, Sawada T, Kato M, Maeno Y, Miyazawa Y, Fujizuka Y, Sekine Y, Koike H, Matsui H, Suzuki K. Fibroblast growth factor receptor inhibitor erdafitinib promotes Mcl-1 degradation and synergistically induces apoptosis with Bcl-xL/Bcl-2 inhibitor in urothelial cancer cells. Biochem Biophys Res Commun 2022; 628:76-83. [DOI: 10.1016/j.bbrc.2022.08.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/12/2022] [Accepted: 08/26/2022] [Indexed: 11/26/2022]
|
24
|
Chiou JT, Lee YC, Wang LJ, Chang LS. BCL2 inhibitor ABT-199 and BCL2L1 inhibitor WEHI-539 coordinately promote NOXA-mediated degradation of MCL1 in human leukemia cells. Chem Biol Interact 2022; 361:109978. [PMID: 35561756 DOI: 10.1016/j.cbi.2022.109978] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023]
Abstract
Human leukemia U937 cells that were continuously treated with hydroquinone (HQ) were transformed into U937/HQ cells with increased MCL1 and BCL2L1 expression. Compared with their parental cells, U937/HQ cells were less sensitive to ABT-263 (BCL2/BCL2L1 inhibitor)/ABT-199 (BCL2 inhibitor) cytotoxicity. The combination of WEHI-539 (BCL2L1 inhibitor) with either ABT-199 or ABT-263 showed synergistic cytotoxicity to U937 and U937/HQ cells. Therefore, we further investigated the cytotoxic mechanism induced by the combination of WEHI-539 and ABT-199. The combined treatment of WEHI-539 and ABT-199 induced NOX4/ROS/p38 MAPK axis-mediated autophagy, which in turn accelerated β-TrCP mRNA turnover. Downregulation of β-TrCP increased Sp1 expression, thereby promoting Sp1-mediated NOXA transcription, which in turn induced NOXA-dependent MCL1 degradation. Enforced expression of MCL1 alleviated the cytotoxicity of WEHI-539 plus ABT-199 to induce the loss of mitochondrial membrane potential and cell viability. WEHI-539 alone induced Sp1/NOXA axis-mediated MCL1 downregulation, while ABT-199 significantly decreased the dose of WEHI-539 by approximately 350- and 50-fold to induce MCL1 suppression in parental and HQ-selected cells, respectively. Furthermore, WEHI-539 sensitized ABT-199-resistant U937 cells to ABT-199 cytotoxicity by inducing NOXA-mediated degradation of MCL1. Collectively, the data in this study indicate that ABT-199 and WEHI-539 cooperatively induce NOXA-dependent MCL1 degradation, and the inhibition of MCL1 mainly explains their combined cytotoxicity in parental, HQ-selected, and ABT-199-resistant U937 cells.
Collapse
Affiliation(s)
- Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
25
|
Mcl-1 levels critically impact the sensitivities of human colorectal cancer cells to APG-1252-M1, a novel Bcl-2/Bcl-XL dual inhibitor that induces Bax-dependent apoptosis. Neoplasia 2022; 29:100798. [PMID: 35462114 PMCID: PMC9046866 DOI: 10.1016/j.neo.2022.100798] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/08/2022] [Accepted: 04/08/2022] [Indexed: 11/23/2022]
Abstract
New treatment options, such as targeted therapies, are urgently needed for the treatment of colorectal cancer (CRC), the third leading cause of cancer-related deaths worldwide. The current study focuses on demonstrating the therapeutic efficacies of APG-1252-M1 (an active form of the prodrug, APG-1252 or pelcitoclax), a highly potent Bcl-2/Bcl-XL dual inhibitor in clinical trials, against CRC and understanding the underlying mechanisms. APG-1252-M1 effectively decreased the survival of CRC cell lines, particularly those expressing relatively low levels of Mcl-1, with the induction of apoptosis. High levels of Mcl-1 were significantly correlated with decreased sensitivity of CRC cell lines to APG-1252-M1. When combined with an Mcl-1 inhibitor, APG-1252-M1 synergistically decreased the survival and induced apoptosis of APG-1252-M1-insensitive cell lines with high levels of Mcl-1. This combination further decreased the survival and enhanced apoptosis even in sensitive cell lines with relatively low levels of Mcl-1, whereas enforced expression of ectopic Mcl-1 in these cells abrogated APG-1252-M1’s effects on decreasing cell survival and inducing apoptosis, which could be reversed by Mcl-1 inhibition. APG-1252-M1 rapidly induced cytochrome C and Smac release from mitochondria with caspase-3 and PARP cleavage. Deficiency of Bax in CRC cells abolished APG-1252-M1’s ability to induce apoptosis, indicating that APG-1252-M1 induces Bax-dependent apoptosis. The current study thus demonstrates the potential of APG-1252-M1 as a monotherapy in the treatment of CRC, particularly those with low Mcl-1 expression, or in combination with an Mcl-1 inhibitor, warranting further evaluation in vivo and in the clinic.
Collapse
|
26
|
Carter BZ, Mak PY, Tao W, Zhang Q, Ruvolo V, Kuruvilla VM, Wang X, Mak DH, Battula VL, Konopleva M, Jabbour EJ, Hughes PE, Chen X, Morrow PK, Andreeff M. Maximal Activation of Apoptosis Signaling by Cotargeting Antiapoptotic Proteins in BH3 Mimetic-Resistant AML and AML Stem Cells. Mol Cancer Ther 2022; 21:879-889. [PMID: 35364607 PMCID: PMC9167707 DOI: 10.1158/1535-7163.mct-21-0690] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/08/2021] [Accepted: 03/18/2022] [Indexed: 11/16/2022]
Abstract
MCL-1 is known to play a major role in resistance to BCL-2 inhibition, but the contribution of other BCL-2 family proteins has not been fully explored. We, here, demonstrate the ineffectiveness of MCL-1 inhibitor AMG176 in venetoclax-resistant, and conversely, of venetoclax in AMG176-resistant acute myelogenous leukemia (AML). Like cells with acquired resistance to venetoclax, cells with acquired resistance to AMG176 express increased MCL-1. Both cells with acquired resistance to venetoclax and to AMG176 express increased levels of BCL-2 and BCL-2A1, decreased BAX, and/or altered levels of other BCL-2 proteins. Cotargeting BCL-2 and MCL-1 was highly synergistic in AML cell lines with intrinsic or acquired resistance to BH3 mimetics or engineered to genetically overexpress BCL-2 or BCL-2A1 or downregulate BAX. The combination effectively eliminated primary AML blasts and stem/progenitor cells resistant to or relapsed after venetoclax-based therapy irrespective of mutations and cytogenetic abnormalities. Venetoclax and AMG176 combination markedly suppressed antiapoptotic BCL-2 proteins and AML stem/progenitor cells and dramatically extended mouse survival (median 336 vs. control 126 days; P < 0.0001) in a patient-derived xenograft (PDX) model developed from a venetoclax/hypomethylating agent therapy-resistant patient with AML. However, decreased BAX levels in the bone marrow residual leukemia cells after 4-week combination treatment may represent a resistance mechanism that contributed to their survival. Enhanced antileukemia activity was also observed in a PDX model of monocytic AML, known to be resistant to venetoclax therapy. Our results support codependence on multiple antiapoptotic BCL-2 proteins and suppression of BAX as mechanisms of AML resistance to individual BH3 mimetics. Cotargeting of MCL-1 and BCL-2 eliminates otherwise apoptosis-resistant cells.
Collapse
Affiliation(s)
- Bing Z. Carter
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Po Yee Mak
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wenjing Tao
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qi Zhang
- Section of Leukemia Biological Research, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vivian Ruvolo
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vinitha M. Kuruvilla
- Section of Leukemia Biological Research, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiangmeng Wang
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Duncan H. Mak
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Venkata L. Battula
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Marina Konopleva
- Section of Leukemia Biological Research, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elias J. Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Xiaoyue Chen
- Oncology Research, Amgen Inc., Thousand Oaks, CA
| | | | - Michael Andreeff
- Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
27
|
Ceramide-induced integrated stress response overcomes Bcl-2 inhibitor resistance in acute myeloid leukemia. Blood 2022; 139:3737-3751. [PMID: 35443029 PMCID: PMC9642852 DOI: 10.1182/blood.2021013277] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 04/04/2022] [Indexed: 11/24/2022] Open
Abstract
Enhancing cellular ceramide levels in AML activates protein kinase R
to induce the integrated stress response. The ISR induces the BH3-only protein Noxa, causing degradation of
Mcl-1 and sensitization of AML to Bcl-2 inhibition.
Inducing cell death by the sphingolipid ceramide is a potential anticancer
strategy, but the underlying mechanisms remain poorly defined. In this study,
triggering an accumulation of ceramide in acute myeloid leukemia (AML) cells by
inhibition of sphingosine kinase induced an apoptotic integrated stress response
(ISR) through protein kinase R–mediated activation of the master
transcription factor ATF4. This effect led to transcription of the BH3-only
protein Noxa and degradation of the prosurvival Mcl-1 protein on which AML cells
are highly dependent for survival. Targeting this novel ISR pathway, in
combination with the Bcl-2 inhibitor venetoclax, synergistically killed primary
AML blasts, including those with venetoclax-resistant mutations, as well as
immunophenotypic leukemic stem cells, and reduced leukemic engraftment in
patient-derived AML xenografts. Collectively, these findings provide mechanistic
insight into the anticancer effects of ceramide and preclinical evidence for new
approaches to augment Bcl-2 inhibition in the therapy of AML and other cancers
with high Mcl-1 dependency.
Collapse
|
28
|
The BCL-2 inhibitor ABT-199/venetoclax synergizes with proteasome inhibition via transactivation of the MCL-1 antagonist NOXA. Cell Death Dis 2022; 8:215. [PMID: 35443750 PMCID: PMC9021261 DOI: 10.1038/s41420-022-01009-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 12/31/2022]
Abstract
Enhanced expression of anti-apoptotic B-cell lymphoma 2 (BCL-2) protein is frequent in cancer. Targeting of BCL-2 with the specific inhibitor ABT-199 (Venetoclax) has significant clinical activity in malignant diseases such as chronic lymphocytic leukemia and multiple myeloma. The small molecule drug ABT-199 mimics the pro-apoptotic BCL-2 homology domain 3 of BH3-only proteins and blocks the hydrophobic BC-groove in BCL-2. We have previously shown that ABT-199 synergizes with the proteasome inhibitor (PI) bortezomib in soft tissue sarcoma derived cells and cell lines to induce apoptosis. Synergistic apoptosis induction relies on the pore-forming effector BAX and expression of the pro-apoptotic BH3-only protein NOXA. Bortezomib augments expression of NOXA by blocking its proteasomal degradation. Interestingly, shown here for the first time, expression of NOXA is strongly enhanced by ABT-199 induced integrated stress response (ISR). ISR transcription factors ATF3 & ATF4 mediate transactivation of the BH3-only protein NOXA which specifically inhibits the anti-apoptotic MCL-1. Thus, NOXA potentiates the efficacy of the BCL-2 inhibitor ABT-199 by simultaneous inhibition of MCL-1. Hence, ABT-199 has a double impact by directly blocking anti-apoptotic BCL-2 and inhibiting MCL-1 via transactivated NOXA. By preventing degradation of NOXA PIs synergize with ABT-199. Synergism of ABT-199 and PIs therefore occurs on several, previously unexpected levels. This finding should prompt clinical evaluation of combinatorial regimens in further malignancies.
Collapse
|
29
|
Bultynck G. Introducing the Special Issue on "Death mechanisms in cellular homeostasis" in honor of Dr. Peter Ruvolo. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119213. [PMID: 34998920 DOI: 10.1016/j.bbamcr.2022.119213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Geert Bultynck
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium.
| |
Collapse
|
30
|
Suraweera CD, Banjara S, Hinds MG, Kvansakul M. Metazoans and Intrinsic Apoptosis: An Evolutionary Analysis of the Bcl-2 Family. Int J Mol Sci 2022; 23:ijms23073691. [PMID: 35409052 PMCID: PMC8998228 DOI: 10.3390/ijms23073691] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 01/12/2023] Open
Abstract
The B-cell lymphoma-2 (Bcl-2) family is a group of genes regulating intrinsic apoptosis, a process controlling events such as development, homeostasis and the innate and adaptive immune responses in metazoans. In higher organisms, Bcl-2 proteins coordinate intrinsic apoptosis through their regulation of the integrity of the mitochondrial outer membrane; this function appears to have originated in the basal metazoans. Bcl-2 genes predate the cnidarian-bilaterian split and have been identified in porifera, placozoans and cnidarians but not ctenophores and some nematodes. The Bcl-2 family is composed of two groups of proteins, one with an α-helical Bcl-2 fold that has been identified in porifera, placozoans, cnidarians, and almost all higher bilaterians. The second group of proteins, the BH3-only group, has little sequence conservation and less well-defined structures and is found in cnidarians and most bilaterians, but not porifera or placozoans. Here we examine the evolutionary relationships between Bcl-2 proteins. We show that the structures of the Bcl-2-fold proteins are highly conserved over evolutionary time. Some metazoans such as the urochordate Oikopleura dioica have lost all Bcl-2 family members. This gene loss indicates that Bcl-2 regulated apoptosis is not an absolute requirement in metazoans, a finding mirrored in recent gene deletion studies in mice. Sequence analysis suggests that at least some Bcl-2 proteins lack the ability to bind BH3-only antagonists and therefore potentially have other non-apoptotic functions. By examining the foundations of the Bcl-2 regulated apoptosis, functional relationships may be clarified that allow us to understand the role of specific Bcl-2 proteins in evolution and disease.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia; (C.D.S.); (S.B.)
| | - Suresh Banjara
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia; (C.D.S.); (S.B.)
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
- Correspondence: (M.G.H.); (M.K.)
| | - Marc Kvansakul
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia; (C.D.S.); (S.B.)
- Correspondence: (M.G.H.); (M.K.)
| |
Collapse
|
31
|
Murthy SRK, Cheng X, Zhuang T, Ly L, Jones O, Basadonna G, Keidar M, Canady J. BCL2A1 regulates Canady Helios Cold Plasma-induced cell death in triple-negative breast cancer. Sci Rep 2022; 12:4038. [PMID: 35260587 PMCID: PMC8904455 DOI: 10.1038/s41598-022-07027-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/09/2022] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is the leading cause of cancer death among women. Triple-negative breast cancer (TNBC) has a poor prognosis and frequently relapses early compared with other subtypes. The Cold Atmospheric Plasma (CAP) is a promising therapy for prognostically poor breast cancer such as TNBC. The Canady Helios Cold Plasma (CHCP) induces cell death in the TNBC cell line without thermal damage, however, the mechanism of cell death by CAP treatment is ambiguous and the mechanism of resistance to cell death in some subset of cells has not been addressed. We investigate the expression profile of 48 apoptotic and 35 oxidative gene markers after CHCP treatment in six different types of breast cancer cell lines including luminal A (ER+ PR+/-HER2-), luminal B (ER+PR+/-HER2+), (ER-PR-HER2+), basal-like: ER-PR-HER2- cells were tested with CHCP at different power settings and at 4 different incubation time. The expression levels of the gene markers were determined at 4 different intervals after the treatment. The protein expression of BCL2A1 was only induced after CHCP treatment in TNBC cell lines (p < 0.01), whereas the HER2-positive and ER, PR positive cell lines showed little or no expression of BCL2A1. The BCL2A1 and TNF-alpha expression levels showed a significant correlation within TNBC cell lines (p < 0.01). Silencing BCL2A1 mRNA by siRNA increased the potency of the CHCP treatment. A Combination of CHCP and CPI203, a BET bromodomain inhibitor, and a BCL2A1 antagonist increased the CHCP-induced cell death (p < 0.05). Our results revealed that BCL2A1 is a key gene for resistance during CHCP induced cell death. This resistance in TNBCs could be reversed with a combination of siRNA or BCL2A1 antagonist-CHCP therapy.
Collapse
Affiliation(s)
- Saravana R K Murthy
- Jerome Canady Research Institute for Advanced Biological and Technological Sciences, Takoma Park, MD, USA
| | - Xiaoqian Cheng
- Jerome Canady Research Institute for Advanced Biological and Technological Sciences, Takoma Park, MD, USA
| | | | - Lawan Ly
- Jerome Canady Research Institute for Advanced Biological and Technological Sciences, Takoma Park, MD, USA
| | - Olivia Jones
- Jerome Canady Research Institute for Advanced Biological and Technological Sciences, Takoma Park, MD, USA
| | | | | | - Jerome Canady
- Jerome Canady Research Institute for Advanced Biological and Technological Sciences, Takoma Park, MD, USA.
- The George Washington University, Washington, DC, USA.
- Holy Cross Hospital, Department of Surgery, Silver Spring, MD, USA.
| |
Collapse
|
32
|
Ramesh P, Di Franco S, Atencia Taboada L, Zhang L, Nicotra A, Stassi G, Medema JP. BCL-XL inhibition induces an FGFR4-mediated rescue response in colorectal cancer. Cell Rep 2022; 38:110374. [PMID: 35172148 DOI: 10.1016/j.celrep.2022.110374] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/27/2021] [Accepted: 01/21/2022] [Indexed: 01/15/2023] Open
Abstract
The heterogeneous therapy response observed in colorectal cancer is in part due to cancer stem cells (CSCs) that resist chemotherapeutic insults. The anti-apoptotic protein BCL-XL plays a critical role in protecting CSCs from cell death, where its inhibition with high doses of BH3 mimetics can induce apoptosis. Here, we screen a compound library for synergy with low-dose BCL-XL inhibitor A-1155463 to identify pathways that regulate sensitivity to BCL-XL inhibition and reveal that fibroblast growth factor receptor (FGFR)4 inhibition effectively sensitizes to A-1155463 both in vitro and in vivo. Mechanistically, we identify a rescue response that is activated upon BCL-XL inhibition and leads to rapid FGF2 secretion and subsequent FGFR4-mediated post-translational stabilization of MCL-1. FGFR4 inhibition prevents MCL-1 upregulation and thereby sensitizes CSCs to BCL-XL inhibition. Altogether, our findings suggest a cell transferable induction of a FGF2/FGFR4 rescue response in CRC that is induced upon BCL-XL inhibition and leads to MCL-1 upregulation.
Collapse
Affiliation(s)
- Prashanthi Ramesh
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Simone Di Franco
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Lidia Atencia Taboada
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Le Zhang
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Annalisa Nicotra
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
33
|
Li L, Li P, Song H, Ma X, Zeng S, Peng Y, Zhang G. Targeting entry into mitochondria for increased anticancer efficacy of BCL-X L-selective inhibitors in lung cancer. Pharmacol Res 2022; 177:106095. [PMID: 35074525 DOI: 10.1016/j.phrs.2022.106095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
The BCL-XL-selective inhibitors exhibit potential clinical application value when combined with chemotherapeutic drugs for the treatment of solid tumors. However, their efficacy in these settings is still low when treated with BCL-XL inhibitors alone in solid tumors. The mechanism responsible for the poor efficacy remains unclear. We show here that unable to interact with target of BCL-XL-selective inhibitors caused by invalid entry into mitochondria is essential for their inefficacy in solid tumors. We demonstrated in non-small-cell lung cancer (NSCLC) cells that the instability of A-1155463 in cells as well as invalid entry into mitochondria of A-1331852, two BCL-XL-selective inhibitors, accounted for their off-target problems. Furthermore, we found that a mitochondria-targeted, non-toxic small molecule NA-2a improved the on-target effect of A-1331852 to enhance its apoptotic regulatory activity, thereby increasing its anticancer activity in NSCLC. Our results indicated that NA-2a was selectively enriched in mitochondria transported by organic-anion-transporting polypeptide (OATP) transporters, which altered the permeability of the mitochondrial membrane, thereby promoting the entrance of A-1331852 to mitochondria and enhancing its disruption of the BIM-BCL-XL complex, which finally led to the increased anticancer activity in vitro and in vivo. Collectively, our data provided overwhelming evidence that the combination of NA-2a and A-1331852 could be used as a promising synergistic therapeutic agent in NSCLC therapy.
Collapse
Affiliation(s)
- Liangping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Pingping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Huanhuan Song
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xuesong Ma
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shulan Zeng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Yan Peng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Guohai Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
34
|
Seyfried F, Stirnweiß FU, Niedermayer A, Enzenmüller S, Hörl RL, Münch V, Köhrer S, Debatin KM, Meyer LH. Synergistic activity of combined inhibition of anti-apoptotic molecules in B-cell precursor ALL. Leukemia 2022; 36:901-912. [PMID: 35031695 PMCID: PMC8979822 DOI: 10.1038/s41375-021-01502-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 12/07/2021] [Accepted: 12/22/2021] [Indexed: 01/01/2023]
Abstract
Targeting BCL-2, a key regulator of survival in B-cell malignancies including precursor B-cell acute lymphoblastic leukemia, has become a promising treatment strategy. However, given the redundancy of anti-apoptotic BCL-2 family proteins (BCL-2, BCL-XL, MCL-1), single targeting may not be sufficient. When analyzing the effects of BH3-mimetics selectively targeting BCL-XL and MCL-1 alone or in combination with the BCL-2 inhibitor venetoclax, heterogeneous sensitivity to either of these inhibitors was found in ALL cell lines and in patient-derived xenografts. Interestingly, some venetoclax-resistant leukemias were sensitive to the MCL-1-selective antagonist S63845 and/or BCL-XL-selective A-1331852 suggesting functional mutual substitution. Consequently, co-inhibition of BCL-2 and MCL-1 or BCL-XL resulted in synergistic apoptosis induction. Functional analysis by BH3-profiling and analysis of protein complexes revealed that venetoclax-treated ALL cells are dependent on MCL-1 and BCL-XL, indicating that MCL-1 or BCL-XL provide an Achilles heel in BCL-2-inhibited cells. The effect of combining BCL-2 and MCL-1 inhibition by venetoclax and S63845 was evaluated in vivo and strongly enhanced anti-leukemia activity was found in a pre-clinical patient-derived xenograft model. Our study offers in-depth molecular analysis of mutual substitution of BCL-2 family proteins in acute lymphoblastic leukemia and provides targets for combination treatment in vivo and in ongoing clinical studies.
Collapse
Affiliation(s)
- Felix Seyfried
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Felix Uli Stirnweiß
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.,International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | - Alexandra Niedermayer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.,International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | - Stefanie Enzenmüller
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Rebecca Louise Hörl
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Vera Münch
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Stefan Köhrer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.,St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Vienna, Austria
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Lüder Hinrich Meyer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
35
|
Diepstraten ST, Anderson MA, Czabotar PE, Lessene G, Strasser A, Kelly GL. The manipulation of apoptosis for cancer therapy using BH3-mimetic drugs. Nat Rev Cancer 2022; 22:45-64. [PMID: 34663943 DOI: 10.1038/s41568-021-00407-4] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/14/2022]
Abstract
Apoptosis is a form of programmed cell death that is regulated by the balance between prosurvival and proapoptotic BCL-2 protein family members. Evasion of apoptosis is a hallmark of cancer that arises when this balance is tipped in favour of survival. One form of anticancer therapeutic, termed 'BH3-mimetic drugs', has been developed to directly activate the apoptosis machinery in malignant cells. These drugs bind to and inhibit specific prosurvival BCL-2 family proteins, thereby mimicking their interaction with the BH3 domains of proapoptotic BCL-2 family proteins. The BCL-2-specific inhibitor venetoclax is approved by the US Food and Drug Administration and many regulatory authorities worldwide for the treatment of chronic lymphocytic leukaemia and acute myeloid leukaemia. BH3-mimetic drugs targeting other BCL-2 prosurvival proteins have been tested in preclinical models of cancer, and drugs targeting MCL-1 or BCL-XL have advanced into phase I clinical trials for certain cancers. As with all therapeutics, efficacy and tolerability need to be carefully balanced to achieve a therapeutic window whereby there is significant anticancer activity with an acceptable safety profile. In this Review, we outline the current state of BH3-mimetic drugs targeting various prosurvival BCL-2 family proteins and discuss emerging data regarding primary and acquired resistance to these agents and approaches that may overcome this. We highlight issues that need to be addressed to further advance the clinical application of BH3-mimetic drugs, both alone and in combination with additional anticancer agents (for example, standard chemotherapeutic drugs or inhibitors of oncogenic kinases), for improved responses in patients with cancer.
Collapse
Affiliation(s)
- Sarah T Diepstraten
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Mary Ann Anderson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Haematology, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Guillaume Lessene
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
36
|
Westaby D, Jimenez-Vacas JM, Padilha A, Varkaris A, Balk SP, de Bono JS, Sharp A. Targeting the Intrinsic Apoptosis Pathway: A Window of Opportunity for Prostate Cancer. Cancers (Basel) 2021; 14:51. [PMID: 35008216 PMCID: PMC8750516 DOI: 10.3390/cancers14010051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
Despite major improvements in the management of advanced prostate cancer over the last 20 years, the disease remains invariably fatal, and new effective therapies are required. The development of novel hormonal agents and taxane chemotherapy has improved outcomes, although primary and acquired resistance remains problematic. Inducing cancer cell death via apoptosis has long been an attractive goal in the treatment of cancer. Apoptosis, a form of regulated cell death, is a highly controlled process, split into two main pathways (intrinsic and extrinsic), and is stimulated by a multitude of factors, including cellular and genotoxic stress. Numerous therapeutic strategies targeting the intrinsic apoptosis pathway are in clinical development, and BH3 mimetics have shown promising efficacy for hematological malignancies. Utilizing these agents for solid malignancies has proved more challenging, though efforts are ongoing. Molecular characterization and the development of predictive biomarkers is likely to be critical for patient selection, by identifying tumors with a vulnerability in the intrinsic apoptosis pathway. This review provides an up-to-date overview of cell death and apoptosis, specifically focusing on the intrinsic pathway. It summarizes the latest approaches for targeting the intrinsic apoptosis pathway with BH3 mimetics and discusses how these strategies may be leveraged to treat prostate cancer.
Collapse
Affiliation(s)
- Daniel Westaby
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| | - Juan M. Jimenez-Vacas
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
| | - Ana Padilha
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
| | - Andreas Varkaris
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (A.V.); (S.P.B.)
| | - Steven P. Balk
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (A.V.); (S.P.B.)
| | - Johann S. de Bono
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| | - Adam Sharp
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| |
Collapse
|
37
|
Ramesh P, Lannagan TRM, Jackstadt R, Atencia Taboada L, Lansu N, Wirapati P, van Hooff SR, Dekker D, Pritchard J, Kirov AB, van Neerven SM, Tejpar S, Kops GJPL, Sansom OJ, Medema JP. BCL-XL is crucial for progression through the adenoma-to-carcinoma sequence of colorectal cancer. Cell Death Differ 2021; 28:3282-3296. [PMID: 34117376 PMCID: PMC8630104 DOI: 10.1038/s41418-021-00816-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Evasion of apoptosis is a hallmark of cancer, which is frequently mediated by upregulation of the antiapoptotic BCL-2 family proteins. In colorectal cancer (CRC), previous work has highlighted differential antiapoptotic protein dependencies determined by the stage of the disease. While intestinal stem cells (ISCs) require BCL-2 for adenoma outgrowth and survival during transformation, ISC-specific MCL1 deletion results in disturbed intestinal homeostasis, eventually contributing to tumorigenesis. Colon cancer stem cells (CSCs), however, no longer require BCL-2 and depend mainly on BCL-XL for their survival. We therefore hypothesized that a shift in antiapoptotic protein reliance occurs in ISCs as the disease progresses from normal to adenoma to carcinoma. By targeting antiapoptotic proteins with specific BH3 mimetics in organoid models of CRC progression, we found that BCL-2 is essential only during ISC transformation while MCL1 inhibition did not affect adenoma outgrowth. BCL-XL, on the other hand, was crucial for stem cell survival throughout the adenoma-to-carcinoma sequence. Furthermore, we identified that the limited window of BCL-2 reliance is a result of its downregulation by miR-17-5p, a microRNA that is upregulated upon APC-mutation driven transformation. Here we show that BCL-XL inhibition effectively impairs adenoma outgrowth in vivo and enhances the efficacy of chemotherapy. In line with this dependency, expression of BCL-XL, but not BCL-2 or MCL1, directly correlated to the outcome of chemotherapy-treated CRC patients. Our results provide insights to enable the rational use of BH3 mimetics in CRC management, particularly underlining the therapeutic potential of BCL-XL targeting mimetics in both early and late-stage disease.
Collapse
Affiliation(s)
- Prashanthi Ramesh
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | | | - Rene Jackstadt
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, G61 1BD, UK
| | - Lidia Atencia Taboada
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Nico Lansu
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Sander R van Hooff
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Danielle Dekker
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jessica Pritchard
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Aleksandar B Kirov
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sanne M van Neerven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sabine Tejpar
- Molecular Digestive Oncology, Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, G61 1QH, UK
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, AmsterdamUMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Porcù E, Benetton M, Bisio V, Da Ros A, Tregnago C, Borella G, Zanon C, Bordi M, Germano G, Manni S, Campello S, Rao DS, Locatelli F, Pigazzi M. The long non-coding RNA CDK6-AS1 overexpression impacts on acute myeloid leukemia differentiation and mitochondrial dynamics. iScience 2021; 24:103350. [PMID: 34816103 PMCID: PMC8591413 DOI: 10.1016/j.isci.2021.103350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/30/2021] [Accepted: 10/22/2021] [Indexed: 12/17/2022] Open
Abstract
Patients with acute myeloid leukemia (AML) carrying high-risk genetic lesions or high residual disease levels after therapy are particularly exposed to the risk of relapse. Here, we identified the long non-coding RNA CDK6-AS1 able to cluster an AML subgroup with peculiar gene signatures linked to hematopoietic cell differentiation and mitochondrial dynamics. CDK6-AS1 silencing triggered hematopoietic commitment in healthy CD34+ cells, whereas in AML cells the pathological undifferentiated state was rescued. This latter phenomenon derived from RUNX1 transcriptional control, responsible for the stemness of hematopoietic precursors and for the block of differentiation in AML. By CDK6-AS1 silencing in vitro, AML mitochondrial mass decreased with augmented pharmacological sensitivity to mitochondria-targeting drugs. In vivo, the combination of tigecycline and cytarabine reduced leukemia progression in the AML-PDX model with high CDK6-AS1 levels, supporting the concept of a mitochondrial vulnerability. Together, these findings uncover CDK6-AS1 as crucial in myeloid differentiation and mitochondrial mass regulation. CDK6-AS1 acts in concert with CDK6 High CDK6-AS1 levels trigger RUNX1 early differentiation arrest in myeloid cells CDK6-AS1 controls mitochondrial mass of AML blasts CDK6-AS1 levels impact on mitochondrial-targeted agents sensitivity
Collapse
Affiliation(s)
- Elena Porcù
- Pediatric Hematology, Oncology and Hematopoietic Cell&Gene Therapy Division of Women's and Children's Health Department, University-Hospital of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy
| | - Maddalena Benetton
- Pediatric Hematology, Oncology and Hematopoietic Cell&Gene Therapy Division of Women's and Children's Health Department, University-Hospital of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy
| | - Valeria Bisio
- Pediatric Hematology, Oncology and Hematopoietic Cell&Gene Therapy Division of Women's and Children's Health Department, University-Hospital of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy
| | - Ambra Da Ros
- Pediatric Hematology, Oncology and Hematopoietic Cell&Gene Therapy Division of Women's and Children's Health Department, University-Hospital of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy
| | - Claudia Tregnago
- Pediatric Hematology, Oncology and Hematopoietic Cell&Gene Therapy Division of Women's and Children's Health Department, University-Hospital of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy
| | - Giulia Borella
- Pediatric Hematology, Oncology and Hematopoietic Cell&Gene Therapy Division of Women's and Children's Health Department, University-Hospital of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy
| | - Carlo Zanon
- Pediatric Onco-Hematology, Stem Cell Transplant and Gene Therapy Laboratory, Istituto di Ricerca Pediatrica - Città della Speranza, 35127 Padova, Italy
| | - Matteo Bordi
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy.,Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, 00143 Rome, Italy
| | - Giuseppe Germano
- Pediatric Onco-Hematology, Stem Cell Transplant and Gene Therapy Laboratory, Istituto di Ricerca Pediatrica - Città della Speranza, 35127 Padova, Italy
| | - Sabrina Manni
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, and Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Silvia Campello
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Sapienza University of Rome, 00165 Roma, Italy
| | - Martina Pigazzi
- Pediatric Hematology, Oncology and Hematopoietic Cell&Gene Therapy Division of Women's and Children's Health Department, University-Hospital of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy.,Pediatric Onco-Hematology, Stem Cell Transplant and Gene Therapy Laboratory, Istituto di Ricerca Pediatrica - Città della Speranza, 35127 Padova, Italy
| |
Collapse
|
39
|
Müller D, Mazzeo P, Koch R, Bösherz MS, Welter S, von Hammerstein-Equord A, Hinterthaner M, Cordes L, Belharazem D, Marx A, Ströbel P, Küffer S. Functional apoptosis profiling identifies MCL-1 and BCL-xL as prognostic markers and therapeutic targets in advanced thymomas and thymic carcinomas. BMC Med 2021; 19:300. [PMID: 34781947 PMCID: PMC8594228 DOI: 10.1186/s12916-021-02158-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Multi-omics studies have shown a high and lack of common driver mutations in most thymomas (TH) and thymic carcinomas (TC) that hamper the development of novel treatment approaches. However, deregulation of apoptosis has been proposed as a common hallmark of TH and TC. BH3 profiling can be utilized to study the readiness of living cancer cells to undergo apoptosis and their dependency on pro-survival BCL-2 family proteins. METHODS We screened a cohort of 62 TH and TC patient samples for expression of BCL-2 family proteins and used the TC cell line 1889c and native TH for dynamic BH3 profiling and treatment with BH3 mimetics. RESULTS Immunohistochemical overexpression of MCL-1 and BCL-xL was a strong prognostic marker of TH and TC, and BH3 profiling indicated a strong dependency on MCL-1 and BCL-xL in TH. Single inhibition of MCL-1 resulted in increased binding of BIM to BCL-xL as an escape mechanism that the combined inhibition of both factors could overcome. Indeed, the inhibition of MCL-1 and BCL-xL in combination induced apoptosis in a caspase-dependent manner in untreated and MCL-1-resistant 1889c cells. CONCLUSION TH and TC are exquisitely dependent on the pro-survival factors MCL-1 and BCL-xL, making them ideal candidates for co-inhibition by BH3 mimetics. Since TH show a heterogeneous dependency on BCL-2 family proteins, upfront BH3 profiling could select patients and tailor the optimal therapy with the least possible toxicity.
Collapse
Affiliation(s)
- Denise Müller
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Paolo Mazzeo
- Department of Haematology and Medical Oncology, University Medical Centre Göttingen, Göttingen, Germany
| | - Raphael Koch
- Department of Haematology and Medical Oncology, University Medical Centre Göttingen, Göttingen, Germany
| | - Mark-Sebastian Bösherz
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Stefan Welter
- Thoracic Surgery Department, Lung Clinic Hemer, Hemer, Germany
| | | | - Marc Hinterthaner
- Department of Thoracic and Cardiovascular Surgery, University Medical Center, Göttingen, Germany
| | - Lucia Cordes
- Thoracic Surgery Department, Lung Clinic Hemer, Hemer, Germany
| | - Djeda Belharazem
- Institute of Pathology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
| | - Stefan Küffer
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| |
Collapse
|
40
|
Rosa N, Ivanova H, Wagner LE, Kale J, La Rovere R, Welkenhuyzen K, Louros N, Karamanou S, Shabardina V, Lemmens I, Vandermarliere E, Hamada K, Ando H, Rousseau F, Schymkowitz J, Tavernier J, Mikoshiba K, Economou A, Andrews DW, Parys JB, Yule DI, Bultynck G. Bcl-xL acts as an inhibitor of IP 3R channels, thereby antagonizing Ca 2+-driven apoptosis. Cell Death Differ 2021; 29:788-805. [PMID: 34750538 PMCID: PMC8990011 DOI: 10.1038/s41418-021-00894-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/09/2022] Open
Abstract
Anti-apoptotic Bcl-2-family members not only act at mitochondria but also at the endoplasmic reticulum, where they impact Ca2+ dynamics by controlling IP3 receptor (IP3R) function. Current models propose distinct roles for Bcl-2 vs. Bcl-xL, with Bcl-2 inhibiting IP3Rs and preventing pro-apoptotic Ca2+ release and Bcl-xL sensitizing IP3Rs to low [IP3] and promoting pro-survival Ca2+ oscillations. We here demonstrate that Bcl-xL too inhibits IP3R-mediated Ca2+ release by interacting with the same IP3R regions as Bcl-2. Via in silico superposition, we previously found that the residue K87 of Bcl-xL spatially resembled K17 of Bcl-2, a residue critical for Bcl-2's IP3R-inhibitory properties. Mutagenesis of K87 in Bcl-xL impaired its binding to IP3R and abrogated Bcl-xL's inhibitory effect on IP3Rs. Single-channel recordings demonstrate that purified Bcl-xL, but not Bcl-xLK87D, suppressed IP3R single-channel openings stimulated by sub-maximal and threshold [IP3]. Moreover, we demonstrate that Bcl-xL-mediated inhibition of IP3Rs contributes to its anti-apoptotic properties against Ca2+-driven apoptosis. Staurosporine (STS) elicits long-lasting Ca2+ elevations in wild-type but not in IP3R-knockout HeLa cells, sensitizing the former to STS treatment. Overexpression of Bcl-xL in wild-type HeLa cells suppressed STS-induced Ca2+ signals and cell death, while Bcl-xLK87D was much less effective in doing so. In the absence of IP3Rs, Bcl-xL and Bcl-xLK87D were equally effective in suppressing STS-induced cell death. Finally, we demonstrate that endogenous Bcl-xL also suppress IP3R activity in MDA-MB-231 breast cancer cells, whereby Bcl-xL knockdown augmented IP3R-mediated Ca2+ release and increased the sensitivity towards STS, without altering the ER Ca2+ content. Hence, this study challenges the current paradigm of divergent functions for Bcl-2 and Bcl-xL in Ca2+-signaling modulation and reveals that, similarly to Bcl-2, Bcl-xL inhibits IP3R-mediated Ca2+ release and IP3R-driven cell death. Our work further underpins that IP3R inhibition is an integral part of Bcl-xL's anti-apoptotic function.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Hristina Ivanova
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Larry E Wagner
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 711, Rochester, NY, 14642, USA
| | - Justin Kale
- Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Rita La Rovere
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Kirsten Welkenhuyzen
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Nikolaos Louros
- VIB Center for Brain and Disease Research, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium.,KU Leuven, Switch Laboratory, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Spyridoula Karamanou
- KU Leuven, Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Campus Gasthuisberg P.O, Box 1037, Herestraat 49, 3000, Leuven, Belgium
| | - Victoria Shabardina
- Institut of Evolutionary Biology, CSIC-Universitat Pompeu Fabra, Passeig Marítim de la Barceloneta 37-49, 08003, Barcelona, Spain
| | - Irma Lemmens
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biomolecular Medicine, Ghent University, and Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | | | - Kozo Hamada
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Hideaki Ando
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, 351-0198, Saitama, Japan
| | - Frederic Rousseau
- VIB Center for Brain and Disease Research, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium.,KU Leuven, Switch Laboratory, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Joost Schymkowitz
- VIB Center for Brain and Disease Research, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium.,KU Leuven, Switch Laboratory, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O/N-1bis Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Jan Tavernier
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biomolecular Medicine, Ghent University, and Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Katsuhiko Mikoshiba
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China.,Faculty of Science, Toho University, Miyama 2-2-1, Funabashi, 274-8510, Chiba, Japan
| | - Anastassios Economou
- KU Leuven, Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Campus Gasthuisberg P.O, Box 1037, Herestraat 49, 3000, Leuven, Belgium
| | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 711, Rochester, NY, 14642, USA
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 Box 802, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
41
|
Lee EF, Fairlie WD. Discovery, development and application of drugs targeting BCL-2 pro-survival proteins in cancer. Biochem Soc Trans 2021; 49:2381-2395. [PMID: 34515749 PMCID: PMC8589430 DOI: 10.1042/bst20210749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
The discovery of a new class of small molecule compounds that target the BCL-2 family of anti-apoptotic proteins is one of the great success stories of basic science leading to translational outcomes in the last 30 years. The eponymous BCL-2 protein was identified over 30 years ago due to its association with cancer. However, it was the unveiling of the biochemistry and structural biology behind it and its close relatives' mechanism(s)-of-action that provided the inspiration for what are now known as 'BH3-mimetics', the first clinically approved drugs designed to specifically inhibit protein-protein interactions. Herein, we chart the history of how these drugs were discovered, their evolution and application in cancer treatment.
Collapse
Affiliation(s)
- Erinna F. Lee
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
- Cell Death and Survival Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia
| | - W. Douglas Fairlie
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
- Cell Death and Survival Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia
| |
Collapse
|
42
|
Fairlie WD, Lee EF. Targeting the BCL-2-regulated apoptotic pathway for the treatment of solid cancers. Biochem Soc Trans 2021; 49:2397-2410. [PMID: 34581776 PMCID: PMC8589438 DOI: 10.1042/bst20210750] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022]
Abstract
The deregulation of apoptosis is a key contributor to tumourigenesis as it can lead to the unwanted survival of rogue cells. Drugs known as the BH3-mimetics targeting the pro-survival members of the BCL-2 protein family to induce apoptosis in cancer cells have achieved clinical success for the treatment of haematological malignancies. However, despite our increasing knowledge of the pro-survival factors mediating the unwanted survival of solid tumour cells, and our growing BH3-mimetics armamentarium, the application of BH3-mimetic therapy in solid cancers has not reached its full potential. This is mainly attributed to the need to identify clinically safe, yet effective, combination strategies to target the multiple pro-survival proteins that typically mediate the survival of solid tumours. In this review, we discuss current and exciting new developments in the field that has the potential to unleash the full power of BH3-mimetic therapy to treat currently recalcitrant solid malignancies.
Collapse
Affiliation(s)
- W. Douglas Fairlie
- Cell Death and Survival Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia
- Cell Death and Survival Laboratory, School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Erinna F. Lee
- Cell Death and Survival Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia
- Cell Death and Survival Laboratory, School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| |
Collapse
|
43
|
Li H, Dong S, Duan L. Difference in the binding mechanisms of ABT-263/43b with Bcl-xL/Bcl-2: computational perspective on the accurate binding free energy analysis. J Mol Model 2021; 27:317. [PMID: 34633547 DOI: 10.1007/s00894-021-04924-9] [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: 03/18/2021] [Accepted: 09/20/2021] [Indexed: 11/26/2022]
Abstract
B-cell lymphoma/leukemia gene-2(Bcl-2) protein family known for regulating cell cycle arrest and subsequent cell death is highly expressed in a variety of cancers. Among them, the Bcl-xL and Bcl-2 are two essential proteins in the Bcl-2 family. In the present work, the differences in binding modes as between the two proteins and two ligands ABT-263/43b were investigated and compared. And the computational alanine scanning combined with the recently developed interaction entropy (AS-IE) method was employed for predicting their binding free energies and finding those amino acids that were more critical during the binding process. The result showed that the binding free energy calculated by the AS-IE method was more in line with experimental values than the molecular mechanics/Poisson-Boltzmann surface area (MM/PBSA) method. Besides, no significant difference was found between Bcl-xL and ABT-263/43b in the binding free energy, which Bcl-xL showed slightly weaker binding free energy to 43b because of the fewer number of key residues with interactions. Nonetheless, compared with the Bcl-2 and 43b complex, the Bcl-2 and ABT-263 system had greater number of key residues interacting with ABT-263, in particular, contribute favorably, resulting in a stronger binding ability for the Bcl-2 and ABT-263 systems. The van der Waals and hydrogen bond contributions were significant in the four protein-ligand complexes. Overall, Tyr108 was found to be the common key residues in the Bcl-xL-ligand complex, while Tyr105, Glu100, and Glu143 were established as the common key residue in the Bcl-2-ligand systems. We hope that the predicted hot spot residues and their energy distributions can guide the design of peptide and small-molecule drugs targeting Bcl-xL and Bcl-2.
Collapse
Affiliation(s)
- Hao Li
- Department of Science and Technology, Shandong Normal University, Jinan, 250014, China
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| | - Shuheng Dong
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| | - Lili Duan
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
44
|
Manzella G, Moonamale DC, Römmele M, Bode P, Wachtel M, Schäfer BW. A combinatorial drug screen in PDX-derived primary rhabdomyosarcoma cells identifies the NOXA - BCL-XL/MCL-1 balance as target for re-sensitization to first-line therapy in recurrent tumors. Neoplasia 2021; 23:929-938. [PMID: 34329950 PMCID: PMC8329430 DOI: 10.1016/j.neo.2021.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/15/2021] [Accepted: 07/02/2021] [Indexed: 01/31/2023] Open
Abstract
First-line therapy for most pediatric sarcoma is based on chemotherapy in combination with radiotherapy and surgery. A significant number of patients experience drug resistance and development of relapsed tumors. Drugs that have the potential to re-sensitize relapsed tumor cells toward chemotherapy treatment are therefore of great clinical interest. Here, we used a drug profiling platform with PDX-derived primary rhabdomyosarcoma cells to screen a large drug library for compounds re-sensitizing relapse tumor cells toward standard chemotherapeutics used in rhabdomyosarcoma therapy. We identified ABT-263 (navitoclax) as most potent compound enhancing general chemosensitivity and used different pharmacologic and genetic approaches in vitro and in vivo to detect the NOXA-BCL-XL/MCL-1 balance to be involved in modulating drug response. Our data therefore suggests that players of the intrinsic mitochondrial apoptotic cascade are major targets for stimulation of response toward first-line therapies in rhabdomyosarcoma.
Collapse
Affiliation(s)
- Gabriele Manzella
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Devmini C Moonamale
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Michaela Römmele
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Peter Bode
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Marco Wachtel
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Beat W Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland.
| |
Collapse
|
45
|
Soliman L, De Souza A, Srinivasan P, Danish M, Bertone P, El-Deiry WS, Carneiro BA. The Role of BCL-2 Proteins in the Development of Castration-resistant Prostate Cancer and Emerging Therapeutic Strategies. Am J Clin Oncol 2021; 44:374-382. [PMID: 34014842 DOI: 10.1097/coc.0000000000000829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The development of androgen resistance in advanced prostate cancer remains a challenging clinical problem. Because androgen deprivation therapy constitutes the backbone of first-line treatments for metastatic prostate cancer, the phenotypic switch from an androgen-dependent to an androgen-independent growth state limits the treatment options for these patients. This critical change from an androgen-dependent to an androgen-independent growth state can be regulated by the B-cell lymphoma gene 2 (BCL-2) family of apoptotic proteins. While the roles of BCL-2 protein family members in the carcinogenesis of prostate cancer have been well-studied, emerging data also delineates their modulation of disease progression to castration-resistant prostate cancer (CRPC). Over the past 2 decades, investigators have sought to describe the mechanisms that underpin this development at the molecular level, yet no recent literature has consolidated these findings in a dedicated review. As new classes of BCL-2 family inhibitors are finding indications for other cancer types, it is time to evaluate how such agents might find stable footing for the treatment of CRPC. Several trials to date have investigated BCL-2 inhibitors as therapeutic agents for CRPC. These therapies include selective BCL-2 inhibitors, pan-BCL-2 inhibitors, and novel inhibitors of MCL-1 and BCL-XL. This review details the research regarding the role of BCL-2 family members in the pathogenesis of prostate cancer and contextualizes these findings within the contemporary landscape of prostate cancer treatment.
Collapse
Affiliation(s)
- Luke Soliman
- Warren Alpert Medical School of Brown University
| | - Andre De Souza
- Warren Alpert Medical School of Brown University
- Division of Hematology/Oncology, Lifespan Cancer Institute
- Cancer Center at Brown University
| | | | - Matthew Danish
- Warren Alpert Medical School of Brown University
- Division of Hematology/Oncology, Lifespan Cancer Institute
| | - Paul Bertone
- Warren Alpert Medical School of Brown University
- Division of Hematology/Oncology, Lifespan Cancer Institute
- Cancer Center at Brown University
| | - Wafik S El-Deiry
- Warren Alpert Medical School of Brown University
- Division of Hematology/Oncology, Lifespan Cancer Institute
- Cancer Center at Brown University
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI
| | - Benedito A Carneiro
- Warren Alpert Medical School of Brown University
- Division of Hematology/Oncology, Lifespan Cancer Institute
- Cancer Center at Brown University
| |
Collapse
|
46
|
Salah HT, DiNardo CD, Konopleva M, Khoury JD. Potential Biomarkers for Treatment Response to the BCL-2 Inhibitor Venetoclax: State of the Art and Future Directions. Cancers (Basel) 2021; 13:2974. [PMID: 34198580 PMCID: PMC8231978 DOI: 10.3390/cancers13122974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/30/2022] Open
Abstract
Intrinsic apoptotic pathway dysregulation plays an essential role in all cancers, particularly hematologic malignancies. This role has led to the development of multiple therapeutic agents targeting this pathway. Venetoclax is a selective BCL-2 inhibitor that has been approved for the treatment of chronic lymphoid leukemia and acute myeloid leukemia. Given the reported resistance to venetoclax, understanding the mechanisms of resistance and the potential biomarkers of response is crucial to ensure optimal drug usage and improved patient outcomes. Mechanisms of resistance to venetoclax include alterations involving the BH3-binding groove, BCL2 gene mutations affecting venetoclax binding, and activation of alternative anti-apoptotic pathways. Moreover, various potential genetic biomarkers of venetoclax resistance have been proposed, including chromosome 17p deletion, trisomy 12, and TP53 loss or mutation. This manuscript provides an overview of biomarkers that could predict treatment response to venetoclax.
Collapse
Affiliation(s)
- Haneen T. Salah
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.D.D.); (M.K.)
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.D.D.); (M.K.)
| | - Joseph D. Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
47
|
Daniels VW, Zoeller JJ, van Gastel N, McQueeney KE, Parvin S, Potter DS, Fell GG, Ferreira VG, Yilma B, Gupta R, Spetz J, Bhola PD, Endress JE, Harris IS, Carrilho E, Sarosiek KA, Scadden DT, Brugge JS, Letai A. Metabolic perturbations sensitize triple-negative breast cancers to apoptosis induced by BH3 mimetics. Sci Signal 2021; 14:14/686/eabc7405. [PMID: 34103421 DOI: 10.1126/scisignal.abc7405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer cells have differential metabolic dependencies compared to their nonmalignant counterparts. However, few metabolism-targeting compounds have been successful in clinical trials. Here, we investigated the metabolic vulnerabilities of triple-negative breast cancer (TNBC), particularly those metabolic perturbations that increased mitochondrial apoptotic priming and sensitivity to BH3 mimetics (drugs that antagonize antiapoptotic proteins). We used high-throughput dynamic BH3 profiling (HT-DBP) to screen a library of metabolism-perturbing small molecules, which revealed inhibitors of the enzyme nicotinamide phosphoribosyltransferase (NAMPT) as top candidates. In some TNBC cells but not in nonmalignant cells, NAMPT inhibitors increased overall apoptotic priming and induced dependencies on specific antiapoptotic BCL-2 family members. Treatment of TNBC cells with NAMPT inhibitors sensitized them to subsequent treatment with BH3 mimetics. The combination of a NAMPT inhibitor (FK866) and an MCL-1 antagonist (S63845) reduced tumor growth in a TNBC patient-derived xenograft model in vivo. We found that NAMPT inhibition reduced NAD+ concentrations below a critical threshold that resulted in depletion of adenine, which was the metabolic trigger that primed TNBC cells for apoptosis. These findings demonstrate a close interaction between metabolic and mitochondrial apoptotic signaling pathways and reveal that exploitation of a tumor-specific metabolic vulnerability can sensitize some TNBC to BH3 mimetics.
Collapse
Affiliation(s)
- Veerle W Daniels
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Ludwig Center at Harvard, Boston, MA 02215, USA
| | - Jason J Zoeller
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Nick van Gastel
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kelley E McQueeney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Salma Parvin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Danielle S Potter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Geoffrey G Fell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Vinícius G Ferreira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos, SP 13568-250, Brazil.,Instituto Nacional de Ciência e Tecnologia de Bioanalítica, INCTBio, Campinas, SP 13083-970, Brazil
| | - Binyam Yilma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Rajat Gupta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Johan Spetz
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - Patrick D Bhola
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jennifer E Endress
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Isaac S Harris
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA.,Department of Biomedical Genetics and Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Emanuel Carrilho
- Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos, SP 13568-250, Brazil.,Instituto Nacional de Ciência e Tecnologia de Bioanalítica, INCTBio, Campinas, SP 13083-970, Brazil
| | - Kristopher A Sarosiek
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joan S Brugge
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. .,Ludwig Center at Harvard, Boston, MA 02215, USA
| |
Collapse
|
48
|
A novel BH3-mimetic, AZD0466, targeting BCL-XL and BCL-2 is effective in pre-clinical models of malignant pleural mesothelioma. Cell Death Discov 2021; 7:122. [PMID: 34050131 PMCID: PMC8163735 DOI: 10.1038/s41420-021-00505-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/15/2021] [Accepted: 05/01/2021] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with treatment limited to Cisplatin and Pemetrexed chemotherapy. Recently, we showed that drugs targeting the BCL-2-regulated apoptosis pathway could kill MPM cell lines in vitro, and control tumor growth in vivo. These studies showed BCL-XL was the dominant pro-survival BCL-2 family member correlating with its high-level expression in cells and patient tumor samples. In this study we show another inhibitor, AZD4320 that targets BCL-XL (and BCL-2), can also potently kill MPM tumor cells in vitro (EC50 values in the 200 nM range) and this effect is enhanced by co-inhibition of MCL-1 using AZD5991. Moreover, we show that a novel nanoparticle, AZD0466, where AZD4320 is chemically conjugated to a PEGylated poly-lysine dendrimer, was as effective as standard-of-care chemotherapy, Cisplatin, at inhibiting tumor growth in mouse xenograft studies, and this effect was enhanced when both drugs were combined. Critically, the degree of thrombocytopenia, an on-target toxicity associated with BCL-XL inhibition, was significantly reduced throughout the treatment period compared to other BCL-XL-targeting BH3-mimetics. These pre-clinical findings provide a rationale for the future clinical evaluation for novel BH3-mimetic formulations in MPM, and indeed, other solid tumor types dependent on BCL-XL.
Collapse
|
49
|
Nguyen W, Lee EF, Evangelista M, Lee M, Harris TJ, Colman PM, Smith NA, Williams LB, Jarman KE, Lowes KN, Haeberli C, Keiser J, Smith BJ, Fairlie WD, Sleebs BE. Optimization of Benzothiazole and Thiazole Hydrazones as Inhibitors of Schistosome BCL-2. ACS Infect Dis 2021; 7:1143-1163. [PMID: 33523649 DOI: 10.1021/acsinfecdis.0c00700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Limited therapeutic options are available for the treatment of human schistosomiasis caused by the parasitic Schistosoma flatworm. The B cell lymphoma-2 (BCL-2)-regulated apoptotic cell death pathway in schistosomes was recently characterized and shown to share similarities with the intrinsic apoptosis pathway in humans. Here, we exploit structural differences in the human and schistosome BCL-2 (sBCL-2) pro-survival proteins toward a novel treatment strategy for schistosomiasis. The benzothiazole hydrazone scaffold previously employed to target human BCL-XL was repurposed as a starting point to target sBCL-2. We utilized X-ray structural data to inform optimization and then applied a scaffold-hop strategy to identify the 5-carboxamide thiazole hydrazone scaffold (43) with potent sBCL-2 activity (IC50 30 nM). Human BCL-XL potency (IC50 13 nM) was inadvertently preserved during the optimization process. The lead analogues from this study exhibit on-target activity in model fibroblast cell lines dependent on either sBCL-2 or human BCL-XL for survival. Further optimization of the thiazole hydrazone class is required to exhibit activity in schistosomes and enhance the potential of this strategy for treating schistosomiasis.
Collapse
Affiliation(s)
- William Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Erinna F. Lee
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne 3086, Australia
| | - Marco Evangelista
- Olivia Newton-John Cancer Research Institute, Heidelberg 3084, Australia
| | - Mihwa Lee
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne 3086, Australia
| | - Tiffany J. Harris
- Olivia Newton-John Cancer Research Institute, Heidelberg 3084, Australia
| | - Peter M. Colman
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Nicholas A. Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne 3086, Australia
| | - Luke B. Williams
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Kate E. Jarman
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Kym N. Lowes
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Cécile Haeberli
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel 4002, Switzerland
- University of Basel, Basel 4001, Switzerland
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel 4002, Switzerland
- University of Basel, Basel 4001, Switzerland
| | - Brian J. Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne 3086, Australia
| | - W. Douglas Fairlie
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne 3086, Australia
| | - Brad E. Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
- Department of Veterinary Biosciences, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
50
|
Zhang L, Ramesh P, Steinmetz M, Medema JP. BH3 Mimetic Sensitivity of Colorectal Cancer Cell Lines in Correlation with Molecular Features Identifies Predictors of Response. Int J Mol Sci 2021; 22:ijms22083811. [PMID: 33917026 PMCID: PMC8067732 DOI: 10.3390/ijms22083811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/27/2021] [Accepted: 04/05/2021] [Indexed: 01/08/2023] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease, which in part explains the differential response to chemotherapy observed in the clinic. BH3 mimetics, which target anti-apoptotic BCL-2 family members, have shown potential in the treatment of hematological malignancies and offer promise for the treatment of solid tumors as well. To gain a comprehensive understanding of the response to BH3 mimetics in CRC and the underlying molecular factors predicting sensitivity, we screened a panel of CRC cell lines with four BH3 mimetics targeting distinct anti-apoptotic BCL-2 proteins. Treatment with compounds alone and in combination revealed potent efficacy of combined MCL-1 and BCL-XL inhibition in inducing CRC cell death, irrespective of molecular features. Importantly, expression of the anti-apoptotic protein target of BH3 mimetics on its own did not predict sensitivity. However, the analysis did identify consensus molecular subtype (CMS) specific response patterns, such as higher resistance to single and combined BCL-2 and MCL-1 inhibition in CMS2 cell lines. Furthermore, analysis of mutation status revealed that KRAS mutant cell lines were more resistant to MCL-1 inhibition. Conclusively, we find that CRC cell lines presented with distinct responses to BH3 mimetics that can in part be predicted by their CMS profile and KRAS/BRAF mutations. Overall, almost all CRC lines share sensitivity in the nanomolar range to combined MCL-1 and BCL-XL targeting suggesting that this would be the preferred approach to target these cancers.
Collapse
Affiliation(s)
- Le Zhang
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.Z.); (P.R.); (M.S.)
- Oncode Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Prashanthi Ramesh
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.Z.); (P.R.); (M.S.)
- Oncode Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Maxime Steinmetz
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.Z.); (P.R.); (M.S.)
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.Z.); (P.R.); (M.S.)
- Oncode Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|