1
|
Wang L, Zhang Q, Ye L, Ye X, Yang W, Zhang H, Zhou X, Ren Y, Ma L, Zhang X, Mei C, Xu G, Li K, Luo Y, Jiang L, Lin P, Zhu S, Lang W, Wang Y, Shen C, Han Y, Liu X, Yang H, Lu C, Sun J, Jin J, Tong H. All-trans retinoic acid enhances the cytotoxic effect of decitabine on myelodysplastic syndromes and acute myeloid leukaemia by activating the RARα-Nrf2 complex. Br J Cancer 2023; 128:691-701. [PMID: 36482192 PMCID: PMC9938271 DOI: 10.1038/s41416-022-02074-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Decitabine (DAC) is used as the first-line therapy in patients with higher-risk myelodysplastic syndromes (HR-MDS) and elderly acute myeloid leukaemia (AML) patients unsuitable for intensive chemotherapy. However, the clinical outcomes of patients treated with DAC as a monotherapy are far from satisfactory. Adding all-trans retinoic acid (ATRA) to DAC reportedly benefitted MDS and elderly AML patients. However, the underlying mechanisms remain unclear and need further explorations from laboratory experiments. METHODS We used MDS and AML cell lines and primary cells to evaluate the combined effects of DAC and ATRA as well as the underlying mechanisms. We used the MOLM-13-luciferase murine xenograft model to verify the enhanced cytotoxic effect of the drug combination. RESULTS The combination treatment reduced the viability of MDS/AML cells in vitro, delayed leukaemia progress, and extended survival in murine xenograft models compared to non- and mono-drug treated models. DAC application as a single agent induced Nrf2 activation and downstream antioxidative response, and restrained reactive oxygen species (ROS) generation, thus leading to DAC resistance. The addition of ATRA blocked Nrf2 activation by activating the RARα-Nrf2 complex, leading to ROS accumulation and ROS-dependent cytotoxicity. CONCLUSIONS These results demonstrate that combining DAC and ATRA has potential for the clinical treatment of HR-MDS/AML and merits further exploration.
Collapse
Affiliation(s)
- Lu Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Qi Zhang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Li Ye
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Xingnong Ye
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Wenli Yang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Hua Zhang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Xinping Zhou
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Yanling Ren
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Liya Ma
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Xiang Zhang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Chen Mei
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Gaixiang Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Kongfei Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Yingwan Luo
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Lingxu Jiang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Peipei Lin
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Department of Radiotherapy, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, Zhejiang, China
| | - Shuanghong Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Wei Lang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Yuxia Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Chuying Shen
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Yueyuan Han
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Xiaozhen Liu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Haiyang Yang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Chenxi Lu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
| | - Jie Sun
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China.
- Myelodysplastic Syndromes Diagnosis and Therapy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China.
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, 310003, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Sobas M, Talarn-Forcadell MC, Martínez-Cuadrón D, Escoda L, García-Pérez MJ, Mariz J, Mela-Osorio MJ, Fernández I, Alonso-Domínguez JM, Cornago-Navascués J, Rodríguez-Macias G, Amutio ME, Rodríguez-Medina C, Esteve J, Sokół A, Murciano-Carrillo T, Calasanz MJ, Barrios M, Barragán E, Sanz MA, Montesinos P. PLZF-RAR α, NPM1-RAR α, and Other Acute Promyelocytic Leukemia Variants: The PETHEMA Registry Experience and Systematic Literature Review. Cancers (Basel) 2020; 12:cancers12051313. [PMID: 32455804 PMCID: PMC7281281 DOI: 10.3390/cancers12051313] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/23/2022] Open
Abstract
It has been suggested that 1–2% of acute promyelocytic leukemia (APL) patients present variant rearrangements of retinoic acid receptor alpha (RARα) fusion gene, with the promyelocytic leukaemia zinc finger (PLZF)/RARα being the most frequent. Resistance to all-trans-retinoic acid (ATRA) and arsenic trioxide (ATO) has been suggested in PLZF/RARα and other variant APLs. Herein, we analyze the incidence, characteristics, and outcomes of variant APLs reported to the multinational PETHEMA (Programa para el Tratamiento de Hemopatias Malignas) registry, and we perform a systematic review in order to shed light on strategies to improve management of these extremely rare diseases. Of 2895 patients with genetically confirmed APL in the PETHEMA registry, 11 had variant APL (0.4%) (9 PLZF-RARα and 2 NPM1-RARα), 9 were men, with median age of 44.6 years (3 months to 76 years), median leucocytes (WBC) 16.8 × 109/L, and frequent coagulopathy. Eight patients were treated with ATRA plus chemotherapy-based regimens, and 3 with chemotherapy-based. As compared to previous reports, complete remission and survival was slightly better in our cohort, with 73% complete remission (CR) and 73% survival despite a high relapse rate (43%). After analyzing our series and performing a comprehensive and critical review of the literature, strong recommendations on appropriate management of variant APL are not possible due to the low number and heterogeneity of patients reported so far.
Collapse
Affiliation(s)
- Marta Sobas
- Blood Neoplasms and Bone Marrow Transplantation, Department of Hematology, Wroclaw Medical University, 50-367 Wrocław, Poland;
| | | | - David Martínez-Cuadrón
- Department of Hematology, Hospital Universitari I Politècnic La Fe, 46-009 Valencia, Spain; (D.M.-C.); (M.A.S.)
- CIBERONC Instituto de Salud Carlos III, 28-020 Madrid, Spain;
| | - Lourdes Escoda
- Hospital of Tarragona “Joan XXIII”, Hematology-ICO, 43-005 Tarragona, Spain; (M.C.T.-F.); (L.E.)
| | | | - Jose Mariz
- Department of Hematology, Istituto Portugues de Oncologi IPO, 4200-072 Porto, Portugal;
| | - María J. Mela-Osorio
- Fundaleu, Department of Hematology, Buenos Aires 1114, Argentina; (M.J.M.-O.); (I.F.)
| | - Isolda Fernández
- Fundaleu, Department of Hematology, Buenos Aires 1114, Argentina; (M.J.M.-O.); (I.F.)
| | - Juan M. Alonso-Domínguez
- Department of Hematology, University Hospital Universitario Fundacion Jimenez Diaz IIS-FJD, 28-040 Madrid, Spain; (J.M.A.-D.); (J.C.-N.)
| | - Javier Cornago-Navascués
- Department of Hematology, University Hospital Universitario Fundacion Jimenez Diaz IIS-FJD, 28-040 Madrid, Spain; (J.M.A.-D.); (J.C.-N.)
| | | | - María E. Amutio
- Department of Hematology, Hospital de Cruces, 48-903 Barakaldo, Spain;
| | - Carlos Rodríguez-Medina
- Department of Hematology, Hospital Universitario Dr. Negrin, 35-010 Las Palmas de Gran Canaria, Spain;
| | - Jordi Esteve
- Department of Hematology, Hospital Clinic, 08-036 Barcelona, Spain;
| | - Agnieszka Sokół
- Department of Paediatric Bone Marrow Transplantation, Oncology and Hematology, Wroclaw Medical University, 50-367 Wrocław, Poland;
| | | | - María J. Calasanz
- Department of Hematology, Clinica Universitaria de Navarra, 31-008 Pamplona, Spain;
| | - Manuel Barrios
- Department of Hematology, Hospital Carlos Haya, 29-014 Málaga, Spain;
| | - Eva Barragán
- CIBERONC Instituto de Salud Carlos III, 28-020 Madrid, Spain;
- Department of Molecular Biology Laboratory, Hospital Universitari I Politècnic La Fe, 46-009 Valencia, Spain
| | - Miguel A. Sanz
- Department of Hematology, Hospital Universitari I Politècnic La Fe, 46-009 Valencia, Spain; (D.M.-C.); (M.A.S.)
- CIBERONC Instituto de Salud Carlos III, 28-020 Madrid, Spain;
| | - Pau Montesinos
- Department of Hematology, Hospital Universitari I Politècnic La Fe, 46-009 Valencia, Spain; (D.M.-C.); (M.A.S.)
- CIBERONC Instituto de Salud Carlos III, 28-020 Madrid, Spain;
- Correspondence:
| |
Collapse
|
3
|
Wang H, Tian L, Liu J, Goldstein A, Bado I, Zhang W, Arenkiel BR, Li Z, Yang M, Du S, Zhao H, Rowley DR, Wong STC, Gugala Z, Zhang XHF. The Osteogenic Niche Is a Calcium Reservoir of Bone Micrometastases and Confers Unexpected Therapeutic Vulnerability. Cancer Cell 2018; 34:823-839.e7. [PMID: 30423299 PMCID: PMC6239211 DOI: 10.1016/j.ccell.2018.10.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 07/10/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023]
Abstract
The fate of disseminated tumor cells is largely determined by microenvironment (ME) niche. The osteogenic niche promotes cancer cell proliferation and bone metastasis progression. We investigated the underlying mechanisms using pre-clinical models and analyses of clinical data. We discovered that the osteogenic niche serves as a calcium (Ca) reservoir for cancer cells through gap junctions. Cancer cells cannot efficiently absorb Ca from ME, but depend on osteogenic cells to increase intracellular Ca concentration. The Ca signaling, together with previously identified mammalian target of rapamycin signaling, promotes bone metastasis progression. Interestingly, effective inhibition of these pathways can be achieved by danusertib, or a combination of everolimus and arsenic trioxide, which provide possibilities of eliminating bone micrometastases using clinically established drugs.
Collapse
Affiliation(s)
- Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Lin Tian
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jun Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Amit Goldstein
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Human and Molecular Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, BCM600, One Baylor Plaza, Houston, TX 77030, USA
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, P.R. China
| | - Meng Yang
- Department of General Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Hong Zhao
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - David R Rowley
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Stephen T C Wong
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Zbigniew Gugala
- Department of Orthopaedic Surgery & Rehabilitation, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, BCM600, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Kasiri S, Shao C, Chen B, Wilson AN, Yenerall P, Timmons BC, Girard L, Tian H, Behrens C, Wistuba II, Gazdar AF, Kim J. GLI1 Blockade Potentiates the Antitumor Activity of PI3K Antagonists in Lung Squamous Cell Carcinoma. Cancer Res 2017; 77:4448-4459. [PMID: 28652248 DOI: 10.1158/0008-5472.can-16-3315] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/13/2017] [Accepted: 06/13/2017] [Indexed: 01/05/2023]
Abstract
Lung squamous cell carcinoma (SCC), strongly associated with smoking, is treated primarily with traditional cytotoxic chemotherapy due to a lack of FDA-approved targeted agents available. Here, we identify the Hedgehog pathway transcription factor GLI1 as a critical driver of lung SCC. Analysis of human lung cancer datasets showed that GLI1 mRNA was highly expressed in human lung SCC and portended a poor prognosis. Inhibition of GLI1 in human lung SCC cell lines suppressed tumor cell clonogenicity and proliferation in culture and in vivo Addition of SHH ligand, SMO antagonists, or other Hedgehog pathway agonists did not affect GLI1 expression in lung SCC cells. However, GLI1 expression was modulated by either inhibition or activation of the PI3K and MAPK pathways. Furthermore, in vivo growth of SCC harboring amplifications of the PI3K gene PIK3CA was attenuated by antagonizing GLI1 and PI3K. Thus, a combinatorial therapeutic strategy that targets the PI3K-mTOR pathway and GLI1 may lead to effective outcomes for PI3K pathway-dependent cancers, in contrast to recent results of human trials with single-agent PI3K antagonists. Cancer Res; 77(16); 4448-59. ©2017 AACR.
Collapse
Affiliation(s)
- Sahba Kasiri
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chunli Shao
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Baozhi Chen
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexandra N Wilson
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Paul Yenerall
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Brenda C Timmons
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Luc Girard
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hui Tian
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Carmen Behrens
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Adi F Gazdar
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Kim
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology Research and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas. .,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
5
|
Mi JQ, Chen SJ, Zhou GB, Yan XJ, Chen Z. Synergistic targeted therapy for acute promyelocytic leukaemia: a model of translational research in human cancer. J Intern Med 2015; 278:627-42. [PMID: 26058416 DOI: 10.1111/joim.12376] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acute promyelocytic leukaemia (APL), the M3 subtype of acute myeloid leukaemia, was once a lethal disease, yet nowadays the majority of patients with APL can be successfully cured by molecularly targeted therapy. This dramatic improvement in the survival rate is an example of the advantage of modern medicine. APL is characterized by a balanced reciprocal chromosomal translocation fusing the promyelocytic leukaemia (PML) gene on chromosome 15 with the retinoic acid receptor α (RARα) gene on chromosome 17. It has been found that all-trans-retinoic acid (ATRA) or arsenic trioxide (ATO) alone exerts therapeutic effect on APL patients with the PML-RARα fusion gene, and the combination of both drugs can act synergistically to further enhance the cure rate of the patients. Here, we provide an insight into the pathogenesis of APL and the mechanisms underlying the respective roles of ATRA and ATO. In addition, treatments that lead to more effective differentiation and apoptosis of APL cells, including leukaemia-initiating cells, and more thorough eradication of the disease will be discussed. Moreover, as a model of translational research, the development of a cure for APL has followed a bidirectional approach of 'bench to bedside' and 'bedside to bench', which can serve as a valuable example for the diagnosis and treatment of other malignancies.
Collapse
Affiliation(s)
- J-Q Mi
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - S-J Chen
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - G-B Zhou
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - X-J Yan
- Department of Hematology, the First Hospital of China Medical University, Shenyang, China
| | - Z Chen
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Yuan B, Yoshino Y, Fukushima H, Markova S, Takagi N, Toyoda H, Kroetz DL. Multidrug resistance-associated protein 4 is a determinant of arsenite resistance. Oncol Rep 2015; 35:147-54. [PMID: 26497925 PMCID: PMC6918809 DOI: 10.3892/or.2015.4343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/19/2015] [Indexed: 12/25/2022] Open
Abstract
Although arsenic trioxide (arsenite, AsIII) has shown a remarkable efficacy in the treatment of acute promyelocytic leukemia patients, multidrug resistance is still a major concern for its clinical use. Multidrug resistance-associated protein 4 (MRP4), which belongs to the ATP-binding cassette (ABC) superfamily of transporters, is localized to the basolateral membrane of hepatocytes and the apical membrane of renal proximal tubule cells. Due to its characteristic localization, MRP4 is proposed as a candidate in the elimination of arsenic and may contribute to resistance to AsIII. To test this hypothesis, stable HEK293 cells overexpressing MRP4 or MRP2 were used to establish the role of these two transporters in AsIII resistance. The IC50 values of AsIII in MRP4 cells were approximately 6-fold higher than those in MRP2 cells, supporting an important role for MRP4 in resistance to AsIII. The capacity of MRP4 to confer resistance to AsIII was further confirmed by a dramatic decrease in the IC50 values with the addition of MK571, an MRP4 inhibitor, and cyclosporine A, a well-known broad-spectrum inhibitor of ABC transporters. Surprisingly, the sensitivity of the MRP2 cells to AsIII was similar to that of the parent cells, although insufficient formation of glutathione and/or Se conjugated arsenic compounds in the MRP2 cells might limit transport. Given that MRP4 is a major contributor to arsenic resistance in vitro, further investigation into the correlation between MRP4 expression and treatment outcome of leukemia patients treated with arsenic-based regimens is warranted.
Collapse
Affiliation(s)
- Bo Yuan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yuta Yoshino
- Department of Clinical Molecular Genetics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hisayo Fukushima
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Svetlana Markova
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hiroo Toyoda
- Department of Clinical Molecular Genetics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
7
|
Chilakapati J, Wallace K, Hernandez-Zavala A, Moore T, Ren H, Kitchin KT. Pharmacokinetic and Genomic Effects of Arsenite in Drinking Water on Mouse Lung in a 30-Day Exposure. Dose Response 2015; 13:1559325815592392. [PMID: 26674514 PMCID: PMC4674186 DOI: 10.1177/1559325815592392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The 2 objectives of this subchronic study were to determine the arsenite drinking water exposure dependent increases in female C3H mouse liver and lung tissue arsenicals and to characterize the dose response (to 0, 0.05, 0.25, 1, 10, and 85 ppm arsenite in drinking water for 30 days and a purified AIN-93M diet) for genomic mouse lung expression patterns. Mouse lungs were analyzed for inorganic arsenic, monomethylated, and dimethylated arsenicals by hydride generation atomic absorption spectroscopy. The total lung mean arsenical levels were 1.4, 22.5, 30.1, 50.9, 105.3, and 316.4 ng/g lung tissue after 0, 0.05, 0.25, 1, 10, and 85 ppm, respectively. At 85 ppm, the total mean lung arsenical levels increased 14-fold and 131-fold when compared to either the lowest noncontrol dose (0.05 ppm) or the control dose, respectively. We found that arsenic exposure elicited minimal numbers of differentially expressed genes (DEGs; 77, 38, 90, 87, and 87 DEGs) after 0.05, 0.25, 1, 10, and 85 ppm, respectively, which were associated with cardiovascular disease, development, differentiation, apoptosis, proliferation, and stress response. After 30 days of arsenite exposure, this study showed monotonic increases in mouse lung arsenical (total arsenic and dimethylarsinic acid) concentrations but no clear dose-related increases in DEG numbers.
Collapse
Affiliation(s)
| | - Kathleen Wallace
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Durham, NC, USA
| | - Araceli Hernandez-Zavala
- Sección de Investigación y Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Tanya Moore
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Durham, NC, USA
| | - Hongzu Ren
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Durham, NC, USA
| | - Kirk T. Kitchin
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Durham, NC, USA
| |
Collapse
|
8
|
Abstract
Key Points
PML/RARA loss or detachment from target promoters suffices to differentiate APL cells. PML/RARA degradation by arsenic thus explains arsenic-induced differentiation.
Collapse
|
9
|
Lao Y, Wang X, Xu N, Zhang H, Xu H. Application of proteomics to determine the mechanism of action of traditional Chinese medicine remedies. JOURNAL OF ETHNOPHARMACOLOGY 2014; 155:1-8. [PMID: 24862488 DOI: 10.1016/j.jep.2014.05.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 05/18/2014] [Accepted: 05/18/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rationale for using traditional Chinese medicine (TCM) is based on the experience that has been gained from its wide use over thousands of years. However, the mechanisms of action of many TCM are still unclear. Proteomics, which mainly characterizes protein functions, protein-protein interactions, and protein modification in tissues or animals, can be used to investigate signaling pathway perturbations in cells or the whole body. Proteomics has improved the discovery process of effective TCM compounds, and has helped to elucidate their possible mechanisms of action. Therefore, a systematic review of the application of proteomics on TCM research is of great importance and necessity. This review strives to describe the literature on the application of proteomics to elucidate the mechanism of action of TCM on various diseases, and provide the essential discussion on the further utilization of proteomics data to accelerate TCM research. MATERIALS AND METHODS Literature survey was performed via electronic search on Pubmed with keywords 'Proteomics' and 'Traditional Chinese Medicine'. The papers written in English were acquired and analyzed in this review. RESULTS This review mainly summarizes the application of proteomics to investigate TCM remedies for neuronal disease, cancer, cardiovascular disease, diabetes, and immunology-related disease. CONCLUSIONS Researchers have applied proteomics to study the mechanism of action of TCM and made substantial progresses. Further studies are required to determine the protein targets of the active compounds, analyze the mechanism of actions in patients, compare the clinical effects with western medicine.
Collapse
Affiliation(s)
- Yuanzhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China
| | - Xiaoyu Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Hongmei Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China.
| |
Collapse
|
10
|
Safa M, Mousavizadeh K, Noori S, Pourfathollah A, Zand H. cAMP protects acute promyelocytic leukemia cells from arsenic trioxide-induced caspase-3 activation and apoptosis. Eur J Pharmacol 2014; 736:115-23. [PMID: 24815320 DOI: 10.1016/j.ejphar.2014.04.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 04/19/2014] [Accepted: 04/23/2014] [Indexed: 11/16/2022]
Abstract
More recently, arsenic trioxide (ATO), was integrated into acute promyelocytic leukemia (APL) treatment, showing high efficacy and tolerability in patients with both ATRA-sensitive and ATRA-resistant APL. ATO could induce apoptosis at relatively high concentrations (0.5 to 2.0 micromol/L) and partial differentiation at low concentrations (0.1 to 0.5 micromol/L) in leukemic promyelocytes. It is known that cAMP agonists enhance low-dose ATO-induced APL cells differentiation. Less well appreciated was the possible interaction between relatively high-doses of ATO and enhanced levels of cAMP in APL cells. Here, we show that elevation of cAMP levels by forskolin inhibited ATO-mediated apoptosis in APL-derived NB4 cells, and this inhibition could be averted by cell permeable cAMP-dependent protein kinase inhibitor (14-22) amide. Inactivating phosphorylation of the proapoptotic protein Bad at Ser118 and phosphorylation of the CREB proto-oncogene at Ser133 were observed upon elevation of cAMP levels in NB4 cells. Phosphorylation of these PKA target proteins is known to promote cell survival in AML cells. The ability of cAMP to endow the APL cells with survival advantage is of particular importance when cAMP agonists may be considered as adjuncts to APL therapy.
Collapse
Affiliation(s)
- Majid Safa
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Kazem Mousavizadeh
- Oncopathology Research Center, and Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Shekoofeh Noori
- Department of Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Pourfathollah
- Department of Medical Laboratory Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Zand
- National Institute and Faculty of Nutrition and Food Technology, Department of Molecular Nutrition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
8-CPT-cAMP/all-trans retinoic acid targets t(11;17) acute promyelocytic leukemia through enhanced cell differentiation and PLZF/RARα degradation. Proc Natl Acad Sci U S A 2013; 110:3495-500. [PMID: 23382200 DOI: 10.1073/pnas.1222863110] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The refractoriness of acute promyelocytic leukemia (APL) with t(11;17)(q23;q21) to all-trans retinoic acid (ATRA)-based therapy concerns clinicians and intrigues basic researchers. By using a murine leukemic model carrying both promyelocytic leukemia zinc finger/retinoic acid receptor-α (PLZF/RARα) and RARα/PLZF fusion genes, we discovered that 8-chlorophenylthio adenosine-3', 5'-cyclic monophosphate (8-CPT-cAMP) enhances cellular differentiation and improves gene trans-activation by ATRA in leukemic blasts. Mechanistically, in combination with ATRA, 8-CPT-cAMP activates PKA, causing phosphorylation of PLZF/RARα at Ser765 and resulting in increased dissociation of the silencing mediator for retinoic acid and thyroid hormone receptors/nuclear receptor corepressor from PLZF/RARα. This process results in changes of local chromatin and transcriptional reactivation of the retinoic acid pathway in leukemic cells. Meanwhile, 8-CPT-cAMP also potentiated ATRA-induced degradation of PLZF/RARα through its Ser765 phosphorylation. In vivo treatment of the t(11;17) APL mouse model demonstrated that 8-CPT-cAMP could significantly improve the therapeutic effect of ATRA by targeting a leukemia-initiating cell activity. This combined therapy, which induces enhanced differentiation and oncoprotein degradation, may benefit t(11;17) APL patients.
Collapse
|
12
|
Chen B, Cao F, Yuan C, Lu X, Shen S, Zhou J, Le XC. Arsenic speciation in saliva of acute promyelocytic leukemia patients undergoing arsenic trioxide treatment. Anal Bioanal Chem 2013; 405:1903-11. [PMID: 23318765 PMCID: PMC3565090 DOI: 10.1007/s00216-012-6700-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/21/2012] [Accepted: 12/22/2012] [Indexed: 11/26/2022]
Abstract
Arsenic trioxide has been successfully used as a therapeutic in the treatment of acute promyelocytic leukemia (APL). Detailed monitoring of the therapeutic arsenic and its metabolites in various accessible specimens of APL patients can contribute to improving treatment efficacy and minimizing arsenic-induced side effects. This article focuses on the determination of arsenic species in saliva samples from APL patients undergoing arsenic treatment. Saliva samples were collected from nine APL patients over three consecutive days. The patients received 10 mg arsenic trioxide each day via intravenous infusion. The saliva samples were analyzed using high-performance liquid chromatography coupled with inductively coupled plasma mass spectrometry. Monomethylarsonous acid and monomethylmonothioarsonic acid were identified along with arsenite, dimethylarsinic acid, monomethylarsonic acid, and arsenate. Arsenite was the predominant arsenic species, accounting for 71.8 % of total arsenic in the saliva. Following the arsenic infusion each day, the percentage of methylated arsenicals significantly decreased, possibly suggesting that the arsenic methylation process was saturated by the high doses immediately after the arsenic infusion. The temporal profiles of arsenic species in saliva following each arsenic infusion over 3 days have provided information on arsenic exposure, metabolism, and excretion. These results suggest that saliva can be used as an appropriate clinical biomarker for monitoring arsenic species in APL patients. Arsenic species and temporal profiles over three days from nine patients ![]()
Collapse
Affiliation(s)
- Baowei Chen
- Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2G3 Canada
| | - Fenglin Cao
- Department of Hematology, Harbin Medical University, Harbin, China
| | - Chungang Yuan
- School of Environmental Sciences and Engineering, North China Electric Power University, Baoding, 071003 Hebei China
| | - Xiufen Lu
- Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2G3 Canada
| | - Shengwen Shen
- Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2G3 Canada
| | - Jin Zhou
- Department of Hematology, Harbin Medical University, Harbin, China
| | - X. Chris Le
- Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2G3 Canada
| |
Collapse
|
13
|
Kim J, Aftab BT, Tang JY, Kim D, Lee AH, Rezaee M, Kim J, Chen B, King EM, Borodovsky A, Riggins GJ, Epstein EH, Beachy PA, Rudin CM. Itraconazole and arsenic trioxide inhibit Hedgehog pathway activation and tumor growth associated with acquired resistance to smoothened antagonists. Cancer Cell 2013; 23:23-34. [PMID: 23291299 PMCID: PMC3548977 DOI: 10.1016/j.ccr.2012.11.017] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 08/27/2012] [Accepted: 11/28/2012] [Indexed: 01/07/2023]
Abstract
Recognition of the multiple roles of Hedgehog signaling in cancer has prompted intensive efforts to develop targeted pathway inhibitors. Leading inhibitors in clinical development act by binding to a common site within Smoothened, a critical pathway component. Acquired Smoothened mutations, including SMO(D477G), confer resistance to these inhibitors. Here, we report that itraconazole and arsenic trioxide, two agents in clinical use that inhibit Hedgehog signaling by mechanisms distinct from that of current Smoothened antagonists, retain inhibitory activity in vitro in the context of all reported resistance-conferring Smoothened mutants and GLI2 overexpression. Itraconazole and arsenic trioxide, alone or in combination, inhibit the growth of medulloblastoma and basal cell carcinoma in vivo, and prolong survival of mice with intracranial drug-resistant SMO(D477G) medulloblastoma.
Collapse
Affiliation(s)
- James Kim
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
- Departments of Biochemistry and of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Blake T. Aftab
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jean Y. Tang
- Department of Dermatology, Stanford University, Stanford, CA 94305, USA
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Daniel Kim
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Alex H. Lee
- Department of Dermatology, Stanford University, Stanford, CA 94305, USA
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Melika Rezaee
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Jynho Kim
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
- Departments of Biochemistry and of Developmental Biology, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Baozhi Chen
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern, Dallas, TX, 75390-8593
| | - Emily M. King
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Alexandra Borodovsky
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Gregory J. Riggins
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ervin H. Epstein
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Philip A. Beachy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
- Departments of Biochemistry and of Developmental Biology, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Corresponding authors: Philip A. Beachy, PhD, Professor of Biochemistry Lokey Stem Cell Research Building, Rm G3120a, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5463, Tel: 650-723-4521, . Charles M. Rudin, MD, PhD, Professor of Oncology, The Johns Hopkins University, Cancer Research Building 2, Room 544, 1550 Orleans Street, Baltimore, MD 21231, Tel: 410-502-0678, Fax: 410-502-0677,
| | - Charles M. Rudin
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Corresponding authors: Philip A. Beachy, PhD, Professor of Biochemistry Lokey Stem Cell Research Building, Rm G3120a, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5463, Tel: 650-723-4521, . Charles M. Rudin, MD, PhD, Professor of Oncology, The Johns Hopkins University, Cancer Research Building 2, Room 544, 1550 Orleans Street, Baltimore, MD 21231, Tel: 410-502-0678, Fax: 410-502-0677,
| |
Collapse
|
14
|
Huai L, Wang C, Zhang C, Li Q, Chen Y, Jia Y, Li Y, Xing H, Tian Z, Rao Q, Wang M, Wang J. Metformin induces differentiation in acute promyelocytic leukemia by activating the MEK/ERK signaling pathway. Biochem Biophys Res Commun 2012; 422:398-404. [PMID: 22575507 DOI: 10.1016/j.bbrc.2012.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 05/01/2012] [Indexed: 12/11/2022]
Abstract
Recent studies have shown that metformin, a widely used antidiabetic agent, may reduce the risk of cancer development. In this study, we investigated the antitumoral effect of metformin on both acute myeloid leukemia (AML) and acute promyelocytic leukemia (APL) cells. Metformin induced apoptosis with partial differentiation in an APL cell line, NB4, but only displayed a proapoptotic effect on several non-M3 AML cell lines. Further analysis revealed that a strong synergistic effect existed between metformin and all-trans retinoic acid (ATRA) during APL cell maturation and that metformin induced the hyperphosphorylation of extracellular signal-regulated kinase (ERK) in APL cells. U0126, a specific MEK/ERK activation inhibitor, abrogated metformin-induced differentiation. Finally, we found that metformin induced the degradation of the oncoproteins PML-RARα and c-Myc and activated caspase-3. In conclusion, these results suggest that metformin treatment may contribute to the enhancement of ATRA-induced differentiation in APL, which may deepen the understanding of APL maturation and thus provide insight for new therapy strategies.
Collapse
Affiliation(s)
- Lei Huai
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Breccia M, Lo-Coco F. Arsenic trioxide for management of acute promyelocytic leukemia: current evidence on its role in front-line therapy and recurrent disease. Expert Opin Pharmacother 2012; 13:1031-43. [PMID: 22468778 DOI: 10.1517/14656566.2012.677436] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Acute promyelocytic leukemia (APL), the most rapidly fatal leukemia only two decades ago, has been converted into the most frequently curable leukemia by the advent of all-trans retinoic acid (ATRA) and its combination with anthracycline-based chemotherapy. More recently, arsenic trioxide (ATO) has been shown to be the most effective single agent in this disease and has been approved for the treatment of relapsed patients both in the United States and Europe. Moreover, ATO has been included in the design of several front-line studies, with the aim to reduce therapy-related toxicity while maintaining the potential of cure. AREAS COVERED First, this review briefly discusses the mechanisms of action and the toxicity profile of ATO. Furthermore, the reported experience on the use of ATO as single agent or in combinatorial schemes both in relapsed and in newly diagnosed patients with APL is critically reviewed. Finally, the use of this agent in special subsets of patients unfit to receive conventional chemotherapy is discussed, along with its potential role in maintenance therapy. EXPERT OPINION While the role of ATO as single agent or in combination with ATRA is well established and recommended by the European LeukemiaNet guidelines as a first option for relapsed patients, the role of the drug in newly diagnosed patients is still uncertain and based only on evidence levels mostly originating from non-randomized trials. The results of ongoing randomized studies should better define the role of ATO in front-line therapy.
Collapse
Affiliation(s)
- Massimo Breccia
- Sapienza University, Department of Cellular Biotechnologies and Hematology, Via Benevento 6, 00161 Rome, Italy.
| | | |
Collapse
|
16
|
Missense mutations in PML-RARA are critical for the lack of responsiveness to arsenic trioxide treatment. Blood 2011; 118:1600-9. [DOI: 10.1182/blood-2011-01-329433] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AbstractArsenic trioxide (As2O3) is a highly effective treatment for patients with refractory/relapsed acute promyelocytic leukemia (APL), but resistance to As2O3 has recently been seen. In the present study, we report the findings that 2 of 15 patients with refractory/relapsed APL treated with As2O3 were clinically As2O3 resistant. Leukemia cells from these 2 patients harbored missense mutations in promyelocytic leukemia gene–retinoic acid receptor-α gene (PML-RARA) transcripts, resulting in amino acid substitutions of A216V and L218P in the PML B2 domain. When wild-type or mutated PML-RARA (PR-WT and PR-B/L-mut, respectively) were overexpressed in HeLa cells, immunoblotting showed SUMOylated and/or oligomerized protein bands in PR-WT but not in PR-B/L-mut after As2O3 treatment. Protein-localization analysis indicated that PR-WT in the soluble fraction was transferred to the insoluble fraction after treatment with As2O3, but PR-B/L-mut was stably detected in fractions both with and without As2O3. Immunofluorescent microscopy analysis showed PR-WT localization as a microgranular pattern in the cytoplasm without As2O3 and as a macrogranular pattern with As2O3. PR-B/L-mut was diffusely observed in the cytoplasm with and without As2O3. Nearly identical localization patterns were observed in patients' primary cells. Therefore, B2 domain mutations may play an important role in aberrant molecular responses to As2O3 and may be critical for As2O3 resistance in APL.
Collapse
|
17
|
Chen SJ, Zhou GB, Zhang XW, Mao JH, de Thé H, Chen Z. From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia. Blood 2011; 117:6425-37. [PMID: 21422471 PMCID: PMC3123014 DOI: 10.1182/blood-2010-11-283598] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Accepted: 03/11/2011] [Indexed: 12/29/2022] Open
Abstract
Arsenic had been used in treating malignancies from the 18th to mid-20th century. In the past 3 decades, arsenic was revived and shown to be able to induce complete remission and to achieve, when combined with all-trans retinoic acid and chemotherapy, a 5-year overall survival of 90% in patients with acute promyelocytic leukemia driven by the t(15;17) translocation-generated promyelocytic leukemia-retinoic acid receptor α (PML-RARα) fusion. Molecularly, arsenic binds thiol residues and induces the formation of reactive oxygen species, thus affecting numerous signaling pathways. Interestingly, arsenic directly binds the C3HC4 zinc finger motif in the RBCC domain of PML and PML-RARα, induces their homodimerization and multimerization, and enhances their interaction with the SUMO E2 conjugase Ubc9, facilitating subsequent sumoylation/ubiquitination and proteasomal degradation. Arsenic-caused intermolecular disulfide formation in PML also contributes to PML-multimerization. All-trans retinoic acid, which targets PML-RARα for degradation through its RARα moiety, synergizes with arsenic in eliminating leukemia-initiating cells. Arsenic perturbs a number of proteins involved in other hematologic malignancies, including chronic myeloid leukemia and adult T-cell leukemia/lymphoma, whereby it may bring new therapeutic benefits. The successful revival of arsenic in acute promyelocytic leukemia, together with modern mechanistic studies, has thus allowed a new paradigm to emerge in translational medicine.
Collapse
Affiliation(s)
- Sai-Juan Chen
- Shanghai Institute of Hematology and State Key Laboratory for Medical Genomics, Rui Jin Hospital/Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, China
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Abstract
As the result of intense clinical and basic research, acute promyelocytic leukemia (APL) has progressively evolved from a deadly to a curable disease. Historically, efforts aimed at understanding the molecular bases for therapy response have repeatedly illuminated APL pathogenesis. The classic model attributes this therapeutic success to the transcriptional reactivation elicited by retinoic acid and the resulting overcoming of the differentiation block characteristic of APL blasts. However, in clinical practice, retinoic acid by itself only rarely yields prolonged remissions, even though it induces massive differentiation. In contrast, as a single agent, arsenic trioxide neither directly activates transcription nor triggers terminal differentiation ex vivo, but cures many patients. Here we review the evidence from recent ex vivo and in vivo studies that allow a reassessment of the role of differentiation in APL cure. We discuss alternative models in which PML-RARA degradation and the subsequent loss of APL cell self-renewal play central roles. Rather than therapy aimed at inducing differentiation, targeting cancer cell self-renewal may represent a more effective goal, achievable by a broader range of therapeutic agents.
Collapse
|
19
|
Wu YL, Zhou HC, Chen GQ. Molecular mechanisms of leukemia-associated protein degradation. ACTA ACUST UNITED AC 2010; 4:363-70. [PMID: 21104160 DOI: 10.1007/s11684-010-0210-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 10/12/2010] [Indexed: 11/25/2022]
Abstract
Chemical biology, using small molecules as probes to study the cellular signaling network, has developed rapidly in recent years. The interaction between chemistry and biology not only provides new insight into the understanding of cellular activities, but also generates new lead compounds for the treatment of diseases. Transcription factors and kinases such as retinoic acid receptor-alpha (RARα), acute myeloid leukemia 1 (AML1), CAAT/enhancer-binding protein α (C/EBPα), c-myc, and c-abl play important roles in the differentiation of hematopoietic stem/progenitor cells. Abnormalities in these proteins may cause the dysregulation of hematopoiesis and even the occurrence of leukemia. Ubiquitin-mediated protein degradation represents a critical mechanism in regulating the cellular levels and functions of these proteins. Thus, targeting protein degradation has been emerging as an important strategy to conquer malignant diseases. In this review, we will summarize the recent advances in the understanding of the roles of protein degradation in leukemia, with an emphasis on the mechanisms revealed by small molecules.
Collapse
MESH Headings
- CCAAT-Enhancer-Binding Protein-alpha/genetics
- CCAAT-Enhancer-Binding Protein-alpha/metabolism
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/metabolism
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/physiopathology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Protein Kinases/genetics
- Protein Kinases/metabolism
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- RUNX1 Translocation Partner 1 Protein
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Ubiquitin/genetics
- Ubiquitin/metabolism
Collapse
Affiliation(s)
- Ying-Li Wu
- Department of Pathophysiology and Chemical Biology Division of Shanghai Universities E-Institutes, Key laboratory of Cell Differentiation and Apoptosis of the Ministry of Education of China, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | | | | |
Collapse
|
20
|
Nasr R, de Thé H. Eradication of acute promyelocytic leukemia-initiating cells by PML/RARA-targeting. Int J Hematol 2010; 91:742-7. [PMID: 20455087 DOI: 10.1007/s12185-010-0582-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2010] [Accepted: 04/19/2010] [Indexed: 12/20/2022]
Abstract
Acute promyelocytic leukemia (APL) is characterized by a t(15;17) translocation that yields a PML/RARA fusion protein. Expression of PML/RARA, a potent transcriptional repressor, induces APL in mice. Both retinoic acid (RA) and arsenic trioxide directly target PML/RARA-mediated transcriptional repression and protein stability, inducing rapid differentiation of the promyelocytes and clinical remission in most APL patients. RA also triggers growth arrest and progressive clearance of leukemia initiating cells (LIC), both ex vivo and in vivo. Suboptimal RA concentrations or expression of the PLZF/RARA variant allows complete RA-induced differentiation, but neither LIC clearance nor disease remission. Thus, RA-induced differentiation and LIC clearance may be uncoupled. The RA/arsenic trioxide association, which dramatically synergizes for PML/RARA degradation but not for differentiation, rapidly clears LIC in a proteasome-dependent manner, resulting in APL eradication in murine models and patients. Collectively, these results demonstrate that LIC clearance, which mirrors PML/RARA degradation, is the primary basis for APL cure by the RA/arsenic trioxide association, rather than differentiation. Oncogene degradation could be a generally applicable therapeutic strategy to clear LICs in several types of tumors.
Collapse
Affiliation(s)
- Rihab Nasr
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
21
|
PML/RARalpha fusion protein transactivates the tissue factor promoter through a GAGC-containing element without direct DNA association. Proc Natl Acad Sci U S A 2010; 107:3716-21. [PMID: 20133705 DOI: 10.1073/pnas.0915006107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A severe coagulopathy is a life-threatening complication of acute promyelocytic leukemia (APL) and is ascribable mainly to the excessive levels of tissue factor (TF) in APL cells regulated in response to the promyelocytic leukemia/retinoic acid receptor alpha (PML/RARalpha) fusion protein. The underlying molecular mechanisms for this regulation remain ill-defined. With U937-PR9 cell lines stably expressing luciferase reporter gene under the control of different mutants of the TF promoter, both luciferase and ChIP data allowed the localization of the PML/RARalpha-responsive sequence in a previously undefined region of the TF promoter at position -230 to -242 devoid of known mammalian transcription factor binding sites. Within this sequence a GAGC motif (-235 to -238) was shown to be crucial because deletion or mutation of these nucleotides impaired both PML/RARalpha interaction and promoter transactivation. However, EMSA results showed that PML/RARalpha did not bind to DNA probes encompassing the -230 to -242 sequences, precluding a direct DNA association. Mutational experiments further suggest that the activator protein 1 (AP-1) sites of the TF promoter are dispensable for PML/RARalpha regulation. This study shows that PML/RARalpha transactivates the TF promoter through an indirect interaction with an element composed of a GAGC motif and the flanking nucleotides, independent of AP-1 binding.
Collapse
|
22
|
Leukemic transformation by the APL fusion protein PRKAR1A-RARα critically depends on recruitment of RXRα. Blood 2010; 115:643-52. [PMID: 19965660 DOI: 10.1182/blood-2009-07-232652] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
PRKAR1A (R1A)–retinoic acid receptor-α (R1A-RARα) is the sixth RARα–containing fusion protein in acute promyelocytic leukemia (APL). Using the murine bone-marrow retroviral transduction/transformation assay, we showed that R1A-RARα fusion protein could transform bone-marrow progenitor/stem cells. In gel-shift assays, R1A-RARα was able to bind to a panel of retinoic acid response elements both as a homodimer and as a heterodimer with RXRα, and demonstrated distinct DNA-binding characteristics compared with wild-type RARα/RXRα or other X-RARα chimeric proteins. The ratio of R1A-RARα to RXRα proteins affected the retinoic acid response element interaction pattern of R1A-RARα/RXRα complexes. Studies comparing R1A-RARα with R1A-RARα(ΔRIIa) demonstrated that the RIIa protein interaction domain located within R1A was responsible for R1A-RARα homodimeric DNA binding and interaction with wild-type R1A protein. However, the RIIa domain was not required for R1A-RARα–mediated transformation because its deletion in R1A-RARα(ΔRIIa) did not compromise its transformation capability. In contrast, introduction of point mutations within the RARα portion of either R1A-RARα or R1A-RARα(ΔRIIa), previously demonstrated to eliminate RXRα interaction or treatment of transduced cells with RXRα shRNA or a RXRα agonist, reduced transformation capability. Thus, leukemic transformation by APL fusion protein PRKAR1A-RARα is critically dependent on RXRα, which suggests RXRα is a promising target for APL.
Collapse
|
23
|
Nasr R, Lallemand-Breitenbach V, Zhu J, Guillemin MC, de Thé H. Therapy-induced PML/RARA proteolysis and acute promyelocytic leukemia cure. Clin Cancer Res 2009; 15:6321-6. [PMID: 19808868 DOI: 10.1158/1078-0432.ccr-09-0209] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Acute promyelocytic leukemia (APL) is characterized by a specific t(15;17) chromosomal translocation that yields the PML/RARA fusion gene. Clinically, besides chemotherapy, two drugs induce clinical remissions: retinoic acid (RA) and arsenic trioxide (As). Both agents directly target PML/RARA-mediated transcriptional repression and protein stability, inducing to various extent promyelocyte differentiation and clinical remission of APL patients. RA targets the RARA moiety of the fusion, whereas arsenic targets its PML part. PML/RARA expression in the mouse is sufficient to initiate APL. The RA-As association, which synergizes for PML/RARA degradation but not for differentiation, rapidly clears leukemia initiating cells (LIC), resulting in APL eradication in murine APL models, but also in several APL clinical trials. Cyclic AMP triggered PML/RARA phosphorylation also enhances RA-induced APL regression, PML/RARA degradation, and LIC clearance, raising new options for therapy-resistant patients. Although differentiation has a major role in debulking of the tumor, PML/RARA degradation seems to be the primary basis for APL eradication by the RA-As association. Oncoprotein degradation could be a general therapeutic strategy that may be extended beyond APL.
Collapse
Affiliation(s)
- Rihab Nasr
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | | | |
Collapse
|
24
|
Erikstein BS, McCormack E, Tronstad KJ, Schwede F, Berge R, Gjertsen BT. Protein kinase A activators and the pan-PPAR agonist tetradecylthioacetic acid elicit synergistic anti-leukaemic effects in AML through CREB. Leuk Res 2009; 34:77-84. [PMID: 19786302 DOI: 10.1016/j.leukres.2009.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 09/05/2009] [Accepted: 09/05/2009] [Indexed: 12/22/2022]
Abstract
Targeting of signal transduction pathways and transcriptional regulation represents an attractive approach for less toxic anti-leukaemic therapy. We combined protein kinase A (PKA) activation with a pan-peroxisome proliferator-activated receptor (PPAR) activator tetradecylthioacetic acid, resulting in synergistic decrease in viability of AML cell lines. PKA isoform II activation appeared to be involved in inhibition of proliferation but not induction of apoptosis in HL-60 cells. Inhibition of CREB function protected against this anti-leukaemic effect with higher efficiency than enforced Bcl-2 expression. Preclinical studies employing the rat AML model Brown Norwegian Myeloid Leukaemia also indicated anti-leukaemic activity of the combination therapy in vivo. In conclusion, combined PKA and pan-PPAR activation should be explored further to determine its therapeutic potential.
Collapse
|
25
|
Nabeshi H, Yoshikawa T, Kamada H, Shibata H, Sugita T, Abe Y, Nagano K, Nomura T, Minowa K, Yamashita T, Itoh N, Yoshioka Y, Tsunoda SI, Tsutsumi Y. Arsenic trioxide inhibits human t cell-lymphotropic virus-1-induced syncytiums by down-regulating gp46. Biol Pharm Bull 2009; 32:1286-8. [PMID: 19571400 DOI: 10.1248/bpb.32.1286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adult T-cell leukemia (ATL) is a severe chemotherapy-resistant malignancy associated with prolonged infection by the human T cell-lymphotropic virus 1 (HTLV-1). One approach to prevent the onset of ATL is to inhibit the growth/transmission of HTLV-1 infected cells using arsenic trioxide (As(2)O(3)). However, there are no reports on the transmission inhibitory effect of As(2)O(3). In this study, we reveal that As(2)O(3) exerts an inhibitory effect on syncytium formation between HTLV-1 infected MT-2 and HeLa cells. In addition, Western blot analysis revealed that the HTLV-1 derived envelope protein gp46 was down regulated by As(2)O(3) treatment, suggesting that As(2)O(3) may inhibit HTLV-1 virus transmission via down-regulation of gp46. These results suggest that As(2)O(3) may be a promising drug to treat refractory HTLV-1-related diseases.
Collapse
Affiliation(s)
- Hiromi Nabeshi
- Laboratory of Pharmaceutical Proteomics, National Institute of Biomedical Innovation
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fu GH, Wang Y, Xi YH, Guo ZW, Liu XB, Bai SZ, Yang BF, Chen GQ. As2O3enhances the anion transport activity of band 3 and the action is related with the C-terminal 16 residues of the protein. J Drug Target 2008; 13:235-43. [PMID: 16051535 DOI: 10.1080/10611860500207060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Successful application of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL) has been attracting worldwide interest, but the exact mechanism for the action of As2O3 remains somewhat obscure. In the present work, we show for the first time that As2O3 facilitates the DIDS-sensitive anion transport activity of band 3 protein in red blood cells (RBCs) isolated from normal adults and APL patients. To elucidate the effect of As2O3 on band 3 protein, constructs encoding the full length of the band 3 transmembrane domain (mdb3) and its C-terminal deletion forms were transfected into yeast cells by a yeast display system. The results demonstrate that deletion of the C-terminal 16 residues of mdb3 (mdb3-d16) does not affect anion transport activity of mdb3 or its sensitivity to DIDS, but decreases its sensitivity to As2O3 in the yeast cell. More intriguingly, the forced expression of intact mdb3 by transfection significantly induces cell apoptosis in HeLa cells, to a higher degree than in cells transfected with mdb3-d16 or empty vector. Expression of activated caspase 3 in HeLa cells also indicates that the C-terminal 16 residues are important for mdb3-mediated apoptosis in cells treated with As2O3. Our results provide the first evidence that As2O3 enhances the anion transport activity of band 3 and the action is related with the C-terminal 16 residues of the protein.
Collapse
Affiliation(s)
- Guo-Hui Fu
- Department of Pathophysiology, Rui-Jin Hospital, Shanghai Second Medical University (SSMU), Shanghai 200025, China.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Li L, Wang J, Ye RD, Shi G, Jin H, Tang X, Yi J. PML/RARalpha fusion protein mediates the unique sensitivity to arsenic cytotoxicity in acute promyelocytic leukemia cells: Mechanisms involve the impairment of cAMP signaling and the aberrant regulation of NADPH oxidase. J Cell Physiol 2008; 217:486-93. [PMID: 18636556 DOI: 10.1002/jcp.21523] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Acute promyelocytic leukemia (APL) cells are characterized by PML/RARalpha fusion protein, high responsiveness to arsenic trioxide (ATO)-induced cytotoxicity and an abundant generation of reactive oxygen species (ROS). In this study we investigated the association among these three features in APL-derived NB4 cells. We found that NADPH oxidase-derived ROS generation was more abundant in NB4 cells compared with monocytic leukemia U937 cells. By using PR9, a sub-line of U937 stably transduced with the inducible PML/RARalpha expression vectors, we attributed disparities on ROS generation and ATO sensitivity to the occurrence of PML/RARalpha fusion protein, since PML/RARalpha-expressing cells appeared higher NADPH oxidase activity, higher ROS level and higher sensitivity to ATO. On the other hand, the basal intensity of cAMP signaling pathway was compared between NB4 and U937 as well as between PR9 cells with or without PML/RARalpha, demonstrating that PML/RARalpha-expressing cells had an impaired cAMP signaling pathway which relieved its inhibitory effect on NADPH oxidase derived ROS generation. In summary, the present study demonstrated the correlation of PML/RARalpha with cAMP signaling pathway, NADPH oxidase and ROS generation in APL cells. PML/RARalpha that bestows NB4 cells various pathological features, paradoxically also endows these cells with the basis for susceptibility to ATO-induced cytotoxcity.
Collapse
Affiliation(s)
- Lingna Li
- Department of Cell Biology, Key Laboratory of The Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciencies, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Acute promyelocytic leukemia (APL) is a distinct subtype of acute myeloid leukemia. Morphologically, it is identified as the M3 subtype of acute myeloid leukemia by the French-American-British classification and cytogenetically is characterized by a balanced reciprocal translocation between chromosomes 15 and 17, which results in the fusion between promyelocytic leukemia (PML) gene and retinoic acid receptor alpha (RARalpha). It seems that the disease is the most malignant form of acute leukemia with a severe bleeding tendency and a fatal course of only weeks. Chemotherapy (CT; daunorubicin, idarubicin and cytosine arabinoside) was the front-line treatment of APL with a complete remission (CR) rate of 75% to 80% in newly diagnosed patients. Despite all these progresses, the median duration of remission ranged from 11 to 25 months and only 35% to 45% of the patients could be cured by CT. Since the introduction of all-trans retinoic acid (ATRA) in the treatment and optimization of the ATRA-based regimens, the CR rate was raised up to 90% to 95% and 5-year disease free survival (DFS) to 74%. The use of arsenic trioxide (ATO) since early 1990s further improved the clinical outcome of refractory or relapsed as well as newly diagnosed APL. In this article, we review the history of introduction of ATRA and ATO into clinical use and the mechanistic studies in understanding this model of cancer targeted therapy.
Collapse
|
29
|
Yang YP, Liang ZQ, Gao B, Jia YL, Qin ZH. Dynamic effects of autophagy on arsenic trioxide-induced death of human leukemia cell line HL60 cells. Acta Pharmacol Sin 2008; 29:123-34. [PMID: 18158874 DOI: 10.1111/j.1745-7254.2008.00732.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIM To evaluate the contribution of an autophagic mechanism to the As2O3- induced death of human acute myeloid leukaemia cell line HL60 cells. METHODS The growth inhibition of HL60 cells induced by As2O3 was assessed with 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide colorimetric assay. The activation of autophagy was determined with monodansylcadaverine labeling and transmission electron microscope. The role of autophagy in the As2O3-induced death of HL60 cells was assessed using autophagic and lysosomal inhibitors. Immunofluorescence, flow cytometry, and Western blot analysis were used to study the apoptotic and autophagic mechanisms. RESULTS After treatment with As2O3, the proliferation of HL60 cells was significantly inhibited and the formation of autophagosomes increased. The blockade of autophagy maturation with the autophagy-specific inhibitor 3-methyladenine (3-MA) or the lysosome-neutralizing agent NH4Cl 1 h before As2O3 potentiated the As2O3-induced death of HL60 cells. In contrast, 3-MA attenuated As2O3-induced death when administered 30 min after As2O3. 3-MA and NH4Cl also inhibited As2O3-induced upregulation of microtubule-associated protein 1 light chain 3, the protein required for autophagy in mammalian cells. Following As2O3, lysosomes were activated as indicated by increased levels of cathepsins B and L. The apoptotic response of HL60 cells to As2O3 was suggested by the collapse of mitochondrial membrane potential, release of cytochrome c from mitochondria, and the activation of caspase-3. Pretreatment with 3-MA prior to As2O3 amplified these apoptotic signals, while posttreatment with 3-MA 30 min after As2O3 attenuated the apoptotic pathways. CONCLUSION Autophagy plays complex roles in the As2O3-induced death of HL60 cells; it inhibits As2O3-induced apoptosis in the initiation stage, but amplifies the As2O3-mediated apoptotic program if it is persistently activated.
Collapse
Affiliation(s)
- Ya-ping Yang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou, China
| | | | | | | | | |
Collapse
|
30
|
Catalano A, Dawson MA, Somana K, Opat S, Schwarer A, Campbell LJ, Iland H. The PRKAR1A gene is fused to RARA in a new variant acute promyelocytic leukemia. Blood 2007; 110:4073-6. [PMID: 17712046 DOI: 10.1182/blood-2007-06-095554] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
We report the molecular and cytogenetic characterization of a novel variant of acute promyelocytic leukemia (APL). The bone marrow showed 88% hypergranular promyelocytes, and the karyotype was 47,XY,+22 [5]/46,XY[30]. Fluorescence in situ hybridization (FISH) indicated disruption and deletion of the 5′-end of the RARA gene. Treatment with all-trans retinoic acid, idarubicin, and arsenic trioxide induced cytogenetic complete remission without morphologic evidence of residual leukemia. The diagnostic marrow was negative for PML-RARA transcripts by reverse transcription–polymerase chain reaction (RT-PCR), but an atypical product was observed. Sequencing showed partial homology to the PRKAR1A gene, encoding the regulatory subunit type I-α of cyclic adenosine monophosphate–dependent protein kinase. RT-PCR using specific primers for PRKAR1A and RARA amplified 2 transcript splice variants of a PRKAR1A-RARA fusion gene, and PRKAR1A and RARA FISH probes confirmed the fusion. This novel PRKAR1A-RARA gene rearrangement is the fifth variant APL in which the RARA partner gene has been identified and the second known rearrangement of PRKAR1A in a malignant disease. This trial was registered at www.actr.org.au with the Australian Clinical Trials Registry as number 12605000070639.
Collapse
MESH Headings
- Aged
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Arsenic Trioxide
- Arsenicals/administration & dosage
- Australia
- Base Sequence/genetics
- Bone Marrow/pathology
- Chromosome Aberrations
- Clinical Trials as Topic
- Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics
- DNA Mutational Analysis
- Humans
- Idarubicin/administration & dosage
- Leukemia, Promyelocytic, Acute/diagnosis
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- Male
- Mutant Chimeric Proteins/genetics
- Neoplasm Proteins/genetics
- Oxides/administration & dosage
- RNA, Messenger/genetics
- Receptors, Retinoic Acid/genetics
- Registries
- Remission Induction
- Retinoic Acid Receptor alpha
- Sequence Deletion/genetics
- Tretinoin/administration & dosage
Collapse
Affiliation(s)
- Alberto Catalano
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, Australia.
| | | | | | | | | | | | | |
Collapse
|
31
|
Zhou GB, Zhang J, Wang ZY, Chen SJ, Chen Z. Treatment of acute promyelocytic leukaemia with all-trans retinoic acid and arsenic trioxide: a paradigm of synergistic molecular targeting therapy. Philos Trans R Soc Lond B Biol Sci 2007; 362:959-71. [PMID: 17317642 PMCID: PMC2435563 DOI: 10.1098/rstb.2007.2026] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
To turn a disease from highly fatal to highly curable is extremely difficult, especially when the disease is a type of cancer. However, we can gain some insight into how this can be done by looking back over the 50-year history of taming acute promyelocytic leukaemia (APL). APL is the M3 type of acute myeloid leukaemia characterized by an accumulation of abnormal promyelocytes in bone marrow, a severe bleeding tendency and the presence of the chromosomal translocation t(15;17) or variants. APL was considered the most fatal type of acute leukaemia five decades ago and the treatment of APL was a nightmare for physicians. Great efforts have been made by scientists worldwide to conquer this disease. The first use of chemotherapy (CT) was unsuccessful due to lack of supportive care and cytotoxic-agent-related exacerbated coagulopathy. The first breakthrough came from the use of anthracyclines which improved the complete remission (CR) rate, though the 5-year overall survival could only be attained in a small proportion of patients. A rational and intriguing hypothesis, to induce differentiation of APL cells rather than killing them, was raised in the 1970s. Laudably, the use of all-trans retinoic acid (ATRA) in treating APL resulted in terminal differentiation of APL cells and a 90-95% CR rate of patients, turning differentiation therapy in cancer treatment from hypothesis to practice. The combination of ATRA with CT further improved the 5-year overall survival. When arsenic trioxide (ATO) was used to treat relapsed APL not only the patients but also the ancient drug were revived. ATO exerts dose-dependent dual effects on APL cells: at low concentration, ATO induces partial differentiation, while at relatively high concentration, it triggers apoptosis. Of note, both ATRA and ATO trigger catabolism of the PML-RARalpha fusion protein which is the key player in APL leukaemogenesis generated from t(15;17), targeting the RARalpha (retinoic acid receptor alpha) or promyelocytic leukaemia (PML) moieties, respectively. Hence, in treating APL both ATRA and ATO represent paradigms for molecularly targeted therapy. At molecular level, ATRA and ATO synergistically modulate multiple downstream pathways/cascades. Strikingly, a clearance of PML-RARalpha transcript in an earlier and more thorough manner, and a higher quality remission and survival in newly diagnosed APL are achieved when ATRA is combined with ATO, as compared to either monotherapy, making APL a curable disease. Thus, the story of APL can serve as a model for the development of curative approaches for disease; it suggests that molecularly synergistic targeted therapies are powerful tools in cancer, and dissection of disease pathogenesis or anatomy of the cancer genome is critical in developing molecular target-based therapies.
Collapse
Affiliation(s)
- Guang-Biao Zhou
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai JiaoTong University School of Medicine (SJTUSM)197, Ruijin Road II, Shanghai 200025, People's Republic of China
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of SciencesGuangzhou Sciences Park, Guangzhou 510663, People's Republic of China
| | - Ji Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai JiaoTong University School of Medicine (SJTUSM)197, Ruijin Road II, Shanghai 200025, People's Republic of China
- Institute of Health Science, SJTUSM and Shanghai Institutes of Biological Sciences, CASSouth Chongqing Road, Shanghai 200025, People's Republic of China
| | - Zhen-Yi Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai JiaoTong University School of Medicine (SJTUSM)197, Ruijin Road II, Shanghai 200025, People's Republic of China
| | - Sai-Juan Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai JiaoTong University School of Medicine (SJTUSM)197, Ruijin Road II, Shanghai 200025, People's Republic of China
| | - Zhu Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai JiaoTong University School of Medicine (SJTUSM)197, Ruijin Road II, Shanghai 200025, People's Republic of China
- Author for correspondence ()
| |
Collapse
|
32
|
Qian W, Liu J, Jin J, Ni W, Xu W. Arsenic trioxide induces not only apoptosis but also autophagic cell death in leukemia cell lines via up-regulation of Beclin-1. Leuk Res 2007; 31:329-39. [PMID: 16882451 DOI: 10.1016/j.leukres.2006.06.021] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 06/05/2006] [Accepted: 06/29/2006] [Indexed: 01/12/2023]
Abstract
Although recent data shows that arsenic trioxide (As2O3) is capable of inducing cell death via cell cycle arrest and apoptosis both in acute promyelocytic leukemia (APL) and in non-APL cells, the mechanisms of As2O3-mediated cell death are not fully understood. In this study, we investigated the in vitro effects of As2O3 on cell growth inhibition and cell death in human T-lymphocytic leukemia and myelodysplastic syndrome (MDS) cell lines. As2O3 significantly inhibited the proliferation of Molt-4 and Mutz-1 cells in dose- and time-dependent manner. Autophagic cell death (programmed cell death type II) and apoptosis (programmed cell death type I) were activated together in leukemia cell lines after exposed to As2O3. Numerous large cytoplasmic inclusions and vacuoles were observed in As2O3-treated cells using electron microscope. Furthermore, 3-methyladenine (an autophagy inhibitor) significantly reduced autophagic cell death and sequentially induced apoptosis. Finally, leukemia cells treated with 4 microM As2O3 showed a considerable up-regulation of Beclin-1 (a Bcl-2-interacting protein) expression, which was independent of transcription of mRNA and required protein synthesis. In addition, Molt-4 cells treated with As2O3 exhibited the down-regulation of Bax protein expression, suggesting that Bax may be involved in accumulating of Beclin-1 and triggering autophagic cell death in As2O3-treated leukemia cells. These results may lead to a better understanding of the mechanism of action of As2O3, and provide a suggestion that As2O3 may be of therapeutic value for the treatment of patients with human T-lymphocytic leukemia and myelodysplastic syndrome.
Collapse
Affiliation(s)
- Wenbin Qian
- Institute of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79# Qingchun Road, Hangzhou 310003, China.
| | | | | | | | | |
Collapse
|
33
|
Vitoux D, Nasr R, de The H. Acute promyelocytic leukemia: New issues on pathogenesis and treatment response. Int J Biochem Cell Biol 2007; 39:1063-70. [PMID: 17468032 DOI: 10.1016/j.biocel.2007.01.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 12/21/2006] [Accepted: 01/01/2007] [Indexed: 11/15/2022]
Abstract
Pathogenesis of acute promyelocytic leukemia appears to be one of the best understood among human malignancies. The ability of retinoic acid (RA) and arsenic trioxide to directly target the oncogenic promyelocytic leukemia-retinoic receptor A (PML-RARA) fusion protein also made this disease the first model for oncogene-targeted therapies. A set of recent data has significantly increased the complexity of our view of acute promyelocytic leukemia pathogenesis, as well as of therapeutic response. This review summarizes and discusses these findings, which yield novels questions and models.
Collapse
MESH Headings
- Arsenic Trioxide
- Arsenicals/pharmacology
- Arsenicals/therapeutic use
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- Models, Biological
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oxides/pharmacology
- Oxides/therapeutic use
- Tretinoin/pharmacology
- Tretinoin/therapeutic use
Collapse
Affiliation(s)
- Dominique Vitoux
- CNRS UMR 7151, Université Paris 7, Equipe labellisée par la Ligue Nationale contre le Cancer, Hôpital Saint-Louis (APHP), 1 av Claude Vellefaux, 75475 Paris Cedex 10, France.
| | | | | |
Collapse
|
34
|
Luo LY, Huang J, Gou BD, Zhang TL, Wang K. Induction of human promyelocytic leukemia HL-60 cell differentiation into monocytes by arsenic sulphide: Involvement of serine/threonine protein phosphatases. Leuk Res 2006; 30:1399-405. [PMID: 16650894 DOI: 10.1016/j.leukres.2006.03.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2005] [Revised: 03/13/2006] [Accepted: 03/13/2006] [Indexed: 10/24/2022]
Abstract
The clinical efficacy of arsenic sulfide (As(4)S(4)), also known as realgar, in the treatment of leukemia in China is prompting people to explore the underlying mechanism. We examined the realgar-induced differentiation in human promyelocytic leukemia cell line HL-60. Cells exhibited proliferation inhibition when treated with 0.10-1.5 microM of realgar, and underwent monocytic differentiation as indicated by morphological changes, NBT reduction assay, and cytofluorometric analyses of the cell surface antigens, CD11b and CD14. Accompanying the differentiation, the activity of serine/threonine protein phosphatase type 1 (PP1) and type 2A (PP2A) were enhanced, whereas the activity of PP2B remained virtually the same compared to the control. When cells were treated with realgar in the presence of an inhibitor of PP1 and 2A or an inhibitor of PP2B, the differentiation of the cells was partially suppressed as revealed by NBT reduction assay and the expression of CD14. Our data demonstrate that realgar induces monocytic differentiation in HL-60 cells and that some serine/threonine protein phosphatases may be involved in the process.
Collapse
Affiliation(s)
- Li-Yun Luo
- Department of Chemical Biology, Peking University School of Pharmaceutical Sciences, 38 Xue Yuan Road, Beijing 100083, PR China
| | | | | | | | | |
Collapse
|
35
|
Lerner A, Epstein P. Cyclic nucleotide phosphodiesterases as targets for treatment of haematological malignancies. Biochem J 2006; 393:21-41. [PMID: 16336197 PMCID: PMC1383661 DOI: 10.1042/bj20051368] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The cAMP signalling pathway has emerged as a key regulator of haematopoietic cell proliferation, differentiation and apoptosis. In parallel, general understanding of the biology of cyclic nucleotide PDEs (phosphodiesterases) has advanced considerably, revealing the remarkable complexity of this enzyme system that regulates the amplitude, kinetics and location of intracellular cAMP-mediated signalling. The development of therapeutic inhibitors of specific PDE gene families has resulted in a growing appreciation of the potential therapeutic application of PDE inhibitors to the treatment of immune-mediated illnesses and haematopoietic malignancies. This review summarizes the expression and function of PDEs in normal haematopoietic cells and the evidence that family-specific inhibitors will be therapeutically useful in myeloid and lymphoid malignancies.
Collapse
Affiliation(s)
- Adam Lerner
- *Evans Department of Medicine, Section of Hematology and Oncology, Boston Medical Center, Boston, MA 02118, U.S.A
- †Department of Pathology, Boston University School of Medicine, Boston, MA 02118, U.S.A
| | - Paul M. Epstein
- ‡Department of Pharmacology, University of Connecticut Health Center, Farmington, CT 06030, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
36
|
Monczor F, Fernandez N, Riveiro E, Mladovan A, Baldi A, Shayo C, Davio C. Histamine H2 receptor overexpression induces U937 cell differentiation despite triggered mechanisms to attenuate cAMP signalling. Biochem Pharmacol 2006; 71:1219-28. [PMID: 16458858 DOI: 10.1016/j.bcp.2005.12.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Revised: 12/26/2005] [Accepted: 12/28/2005] [Indexed: 10/25/2022]
Abstract
Knowing that cell-surface receptors that recognize and respond to extracellular stimuli are key components for the regular communication between individual cells required for the survival of any living organism, the aim of the present work was to investigate the effect of H2R overexpression on the U937 signal transduction pathway and its consequences on cell proliferation and differentiation. The overexpression of H2R led to an increase in cAMP basal levels, a leftward shift of agonist concentration-response curves, and similar maximal response to agonist treatment, suggesting that overexpressed H2Rs act as functional spare receptors. In this system cells triggered several mechanisms tending to restore cAMP basal levels to those of the naïve cells. H2R overexpression induced PDE activity stimulation and GRK2 overexpression. In spite of the onset of these regulatory mechanisms, H2 agonist and rolipram treatments induced the terminal differentiation of the H2R overexpressed clone, conversely to the naïve cells. Present findings show that stably H2R overexpression alters cAMP signalling as the result of not only the amounts of second messenger generated but also the activation or upregulation of various components of signalling cascade, leading to an adapted biologically unique system. This adaptation may represent an advantage or a disadvantage, depending on the biological system, but in any case, the existence of compensatory mechanisms should be considered when a clinical treatment is designed.
Collapse
Affiliation(s)
- Federico Monczor
- Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
37
|
Hwang DR, Tsai YC, Lee JC, Huang KK, Lin RK, Ho CH, Chiou JM, Lin YT, Hsu JTA, Yeh CT. Inhibition of hepatitis C virus replication by arsenic trioxide. Antimicrob Agents Chemother 2004; 48:2876-82. [PMID: 15273095 PMCID: PMC478516 DOI: 10.1128/aac.48.8.2876-2882.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) is a serious global problem, and present therapeutics are inadequate to cure HCV infection. In the present study, various antiviral assays show that As2O3 at submicromolar concentrations is capable of inhibiting HCV replication. The 50% effective concentration (EC50) of As2O3 required to inhibit HCV replication was 0.35 microM when it was determined by a reporter-based HCV replication assay, and the EC50 was below 0.2 microM when it was determined by quantitative reverse transcription-PCR analysis. As2O3 did not cause cellular toxicity at this concentration, as revealed by an MTS [3-(4,5-dimethylthiozol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt] assay. A combination of As2O3 and alpha interferon exerted synergistic effects against HCV, as revealed by a multiple linear logistic model and isobologram analysis. Furthermore, in an alternative HCV antiviral system that may recapitulate additional steps involved in HCV infection and replication, As2O3 at 0.3 microM totally abolished the HCV signal, whereas alpha interferon at a high dose (5,000 IU/ml) only partially suppressed the HCV signal. The study highlights the indications for use of a novel class of anti-HCV agent. Further elucidation of the exact antiviral mechanism of As2O3 may lead to the development of agents with potent activities against HCV or related viruses.
Collapse
Affiliation(s)
- Der-Ren Hwang
- Division of Biotechnology & Pharmaceutical Research, National Health Research Institutes, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kamashev D, Vitoux D, De Thé H. PML-RARA-RXR oligomers mediate retinoid and rexinoid/cAMP cross-talk in acute promyelocytic leukemia cell differentiation. ACTA ACUST UNITED AC 2004; 199:1163-74. [PMID: 15096541 PMCID: PMC2211888 DOI: 10.1084/jem.20032226] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
PML-RARA was proposed to initiate acute promyelocytic leukemia (APL) through PML-RARA homodimer-triggered repression. Here, we examined the nature of the PML-RARA protein complex and of its DNA targets in APL cells. Using a selection/amplification approach, we demonstrate that PML-RARA targets consist of two AGGTCA elements in an astonishing variety of orientations and spacings, pointing to highly relaxed structural constrains for DNA binding and identifying a major gain of function of this oncogene. PML-RARA-specific response elements were identified, which all conveyed a major transcriptional response to RA only in APL cells. In these cells, we demonstrate that PML-RARA oligomers are complexed to RXR. Directly probing PML-RARA function in APL cells, we found that the differentiation enhancer cyclic AMP (cAMP) boosted transcriptional activation by RA. cAMP also reversed the normal silencing (subordination) of the transactivating function of RXR when bound to RARA or PML-RARA, demonstrating that the alternate rexinoid/cAMP-triggered APL differentiation pathway also activates PML-RARA targets. Finally, cAMP restored both RA-triggered differentiation and PML-RARA transcriptional activation in mutant RA-resistant APL cells. Collectively, our findings directly demonstrate that APL cell differentiation parallels transcriptional activation through PML-RARA-RXR oligomers and that those are functionally targeted by cAMP, identifying this agent as another oncogene-targeted therapy.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites
- COS Cells
- Cell Differentiation
- Cell Line
- Cyclic AMP/metabolism
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- Humans
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Neoplasm Proteins/chemistry
- Neoplasm Proteins/metabolism
- Nuclear Proteins/chemistry
- Nuclear Proteins/metabolism
- Promyelocytic Leukemia Protein
- Receptor Cross-Talk
- Receptors, Retinoic Acid/chemistry
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
- Retinoid X Receptors
- Retinoids/metabolism
- Transcription Factors/chemistry
- Transcription Factors/metabolism
- Transcriptional Activation
- Tumor Suppressor Proteins
- U937 Cells
Collapse
Affiliation(s)
- Dmitrii Kamashev
- CNRS UPR9051, Hôpital St. Louis, Laboratoire associé N degrees 11, 1, Av. C. Vellefaux, 75475 Paris, Cedex 10, France.
| | | | | |
Collapse
|
39
|
Shen ZX, Shi ZZ, Fang J, Gu BW, Li JM, Zhu YM, Shi JY, Zheng PZ, Yan H, Liu YF, Chen Y, Shen Y, Wu W, Tang W, Waxman S, De Thé H, Wang ZY, Chen SJ, Chen Z. All-trans retinoic acid/As2O3 combination yields a high quality remission and survival in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci U S A 2004; 101:5328-35. [PMID: 15044693 PMCID: PMC397380 DOI: 10.1073/pnas.0400053101] [Citation(s) in RCA: 455] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Both all-trans retinoic acid (ATRA) and arsenic trioxide (As(2)O(3)) have proven to be very effective in obtaining high clinical complete remission (CR) rates in acute promyelocytic leukemia (APL), but they had not been used jointly in an integrated treatment protocol for remission induction or maintenance among newly diagnosed APL patients. In this study, 61 newly diagnosed APL subjects were randomized into three treatment groups, namely by ATRA, As(2)O(3), and the combination of the two drugs. CR was determined by hematological analysis, tumor burden was examined with real-time quantitative RT-PCR of the PML-RAR alpha (promyelocytic leukemia-retinoic acid receptor alpha) fusion transcripts, and side effects were evaluated by means of clinical examinations. Mechanisms possibly involved were also investigated with cellular and molecular biology methods. Although CR rates in three groups were all high (> or =90%), the time to achieve CR differed significantly, with that of the combination group being the shortest one. Earlier recovery of platelet count was also found in this group. The disease burden as reflected by fold change of PML-RAR alpha transcripts at CR decreased more significantly in combined therapy as compared with ATRA or As(2)O(3) mono-therapy (P < 0.01). This difference persisted after consolidation (P < 0.05). Importantly, all 20 cases in the combination group remained in CR whereas 7 of 37 cases treated with mono-therapy relapsed (P < 0.05) after a follow-up of 8-30 months (median: 18 months). Synergism of ATRA and As(2)O(3) on apoptosis and degradation of PML-RAR alpha oncoprotein might provide a plausible explanation for superior efficacy of combination therapy in clinic. In conclusion, the ATRA/As(2)O(3) combination for remission/maintenance therapy of APL brings much better results than either of the two drugs used alone in terms of the quality of CR and the status of the disease-free survival.
Collapse
Affiliation(s)
- Zhi-Xiang Shen
- Shanghai Institute of Hematology, State Key Lab of Medical Genomics, Rui Jin Hospital, Shanghai Second Medical University, 197 Rui Jin Road II, Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shayo C, Legnazzi BL, Monczor F, Fernández N, Riveiro ME, Baldi A, Davio C. The time-course of cyclic AMP signaling is critical for leukemia U-937 cell differentiation. Biochem Biophys Res Commun 2004; 314:798-804. [PMID: 14741706 DOI: 10.1016/j.bbrc.2003.12.166] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The regulation of the cAMP signaling is intimately involved in several cellular processes, including cell differentiation. Here, we provide strong evidence supporting that the time-course of cAMP signal is critical for leukemia U-937 cell differentiation. Three stimulating-cAMP agents were used to analyze the correlation between cAMP time-course and cell differentiation. All three agents denoted similar cAMP maximal responses in dose-response experiments. The kinetic of desensitization showed differential characteristics, while H2 receptor desensitized homologously without affecting PGE2 or forskolin effect, PGE2 response showed mixed desensitization characterized by a homologous initial phase followed by a heterologous phase. Regarding forskolin, long-term stimuli attenuated PGE2 and H2 agonist response without affecting adenylyl cyclase activity. In the absence of phosphodiesterase inhibitors, the three agents induced similar maximal cAMP levels after 5 min, but only that induced by the H2 agonist returned to basal levels. Consistent with this observation, H2 agonist was not able to induce U-937 cell maturation in contrast to PGE2 and forskolin, supporting the importance of time-course signaling in the determination of cell behavior.
Collapse
Affiliation(s)
- Carina Shayo
- Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
41
|
Santibáñez JF, Olivares D, Guerrero J, Martínez J. Cyclic AMP inhibits TGFbeta1-induced cell-scattering and invasiveness in murine-transformed keratinocytes. Int J Cancer 2004; 107:715-20. [PMID: 14566820 DOI: 10.1002/ijc.11457] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mouse-transformed keratinocytes cultured in the presence of transforming growth factor beta1 (TGF-beta1) acquire an array of morphologic and functional properties that give rise to a migratory phenotype that expresses mesenchymal molecular markers. This cellular conversion involves activation of the Ras-ERK pathway, enhancement of urokinase (uPA) and matrix metalloproteinase-9 (MMP-9) expression and induction of invasiveness. In our present work, we demonstrate that cAMP and forskolin are able to prevent the expression of these mesenchymal properties, probably due to blockade of the Ras-ERK pathway. Our results also show that cAMP and forskolin are able to abolish the TGF-beta1-induced reorganization of the actin cytoskeleton that is characteristic of the mesenchymal phenotype and also inhibits the disruption of the E-cadherin cell to cell interactions. The latter responses seem to depend on the activity of protein kinase A, as demonstrated by the activation of the Ras-ERK pathway by specific protein kinase A inhibitors.
Collapse
Affiliation(s)
- Juan F Santibáñez
- Laboratorio de Biología Celular INTA, Universidad de Chile, Santiago, Chile.
| | | | | | | |
Collapse
|
42
|
|
43
|
Mistry AR, Pedersen EW, Solomon E, Grimwade D. The molecular pathogenesis of acute promyelocytic leukaemia: implications for the clinical management of the disease. Blood Rev 2003; 17:71-97. [PMID: 12642121 DOI: 10.1016/s0268-960x(02)00075-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Acute promyelocytic leukaemia (APL) is characterised by chromosomal rearrangements of 17q21, leading to fusion of the gene encoding retinoic acid receptor alpha (RARalpha) to a number of alternative partner genes (X), the most frequent of which are PML (>95%), PLZF (0.8%) and NPM (0.5%). Over the last few years, it has been established that the X-RARalpha fusion proteins play a key role in the pathogenesis of APL through recruitment of co-repressors and the histone deacetylase (HDAC)-complex to repress genes implicated in myeloid differentiation. Paradoxically, the X-RARalpha fusion protein has the potential to mediate myeloid differentiation at pharmacological doses of its ligand (all trans-retinoic acid (ATRA)), which is dependent on the dissociation of the HDAC/co-repressor complex. Arsenic compounds have also been shown to be promising therapeutic agents, leading to differentiation and apoptosis of APL blasts. It is now apparent that the nature of the RARalpha-fusion partner is a critical determinant of response to ATRA and arsenic, underlining the importance of cytogenetic and molecular characterisation of patients with suspected APL to determine the most appropriate treatment approach. Standard protocols involving ATRA combined with anthracycline-based chemotherapy, lead to cure of approximately 70% patients with PML-RARalpha-associated APL. Patients at high risk of relapse can be identified by minimal residual disease monitoring. The challenge for future studies is to improve complete remission rates through reduction of induction deaths, particularly due to haemorrhage, identification of patients at high risk of relapse who would benefit from additional therapy, and identification of a favourable-risk group, for which treatment intensity could be reduced, thereby reducing risks of treatment toxicity and development of secondary leukaemia/myelodysplasia. With the advent of ATRA and arsenic, APL has already provided the first example of successful molecularly targeted therapy; it is hoped that with further understanding of the pathogenesis of the disease, the next decade will yield further improvements in the outlook for these patients.
Collapse
MESH Headings
- Animals
- Gene Rearrangement
- Humans
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- Leukemia, Promyelocytic, Acute/therapy
- Models, Biological
- Mutation
- Neoplasm, Residual/genetics
- Neoplasm, Residual/pathology
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
- Translocation, Genetic
Collapse
Affiliation(s)
- Anita R Mistry
- Division of Medical and Molecular Genetics, Guy's, King's and St Thomas' School of Medicine, London, UK
| | | | | | | |
Collapse
|
44
|
Abstract
Conventional treatment of acute leukemia involves the use of cytotoxic agents (chemotherapy), but other strategies have been explored. All-trans retinoic acid (ATRA) and arsenic have clearly been effective in the treatment of acute promyelocytic leukemia (APL), which creates the possibility that other types of acute leukemia can be conquered by selectively inducing differentiation and/or apoptosis. A great number of investigations have been performed to elucidate the mechanisms and search for effective agents in the treatment of other types of acute leukemia by these new strategies. Progress at the molecular level has been achieved in explaining the mechanisms of action of ATRA and arsenic compounds, and several new agents have emerged, although their clinical effectiveness remains to be confirmed. Mechanism-/gene-based targeted therapy and a combination of different strategies will improve the treatment of acute leukemia.
Collapse
Affiliation(s)
- Zhen-Yi Wang
- Shanghai Second Medical University, Shangai Institute of Hematology, Shanghai, China
| |
Collapse
|
45
|
Abstract
Abstract
Conventional treatment of acute leukemia involves the use of cytotoxic agents (chemotherapy), but other strategies have been explored. All-trans retinoic acid (ATRA) and arsenic have clearly been effective in the treatment of acute promyelocytic leukemia (APL), which creates the possibility that other types of acute leukemia can be conquered by selectively inducing differentiation and/or apoptosis. A great number of investigations have been performed to elucidate the mechanisms and search for effective agents in the treatment of other types of acute leukemia by these new strategies. Progress at the molecular level has been achieved in explaining the mechanisms of action of ATRA and arsenic compounds, and several new agents have emerged, although their clinical effectiveness remains to be confirmed. Mechanism-/gene-based targeted therapy and a combination of different strategies will improve the treatment of acute leukemia.
Collapse
|
46
|
Guillemin MC, Raffoux E, Vitoux D, Kogan S, Soilihi H, Lallemand-Breitenbach V, Zhu J, Janin A, Daniel MT, Gourmel B, Degos L, Dombret H, Lanotte M, De Thé H. In vivo activation of cAMP signaling induces growth arrest and differentiation in acute promyelocytic leukemia. J Exp Med 2002; 196:1373-80. [PMID: 12438428 PMCID: PMC2193985 DOI: 10.1084/jem.20021129] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Differentiation therapy for acute myeloid leukemia uses transcriptional modulators to reprogram cancer cells. The most relevant clinical example is acute promyelocytic leukemia (APL), which responds dramatically to either retinoic acid (RA) or arsenic trioxide (As(2)O(3)). In many myeloid leukemia cell lines, cyclic adenosine monophosphate (cAMP) triggers growth arrest, cell death, or differentiation, often in synergy with RA. Nevertheless, the toxicity of cAMP derivatives and lack of suitable models has hampered trials designed to assess the in vivo relevance of theses observations. We show that, in an APL cell line, cAMP analogs blocked cell growth and unraveled As(2)O(3)-triggered differentiation. Similarly, in RA-sensitive or RA-resistant mouse models of APL, continuous infusions of 8-chloro-cyclic adenosine monophosphate (8-Cl-cAMP) triggered major growth arrest, greatly enhanced both spontaneous and RA- or As(2)O(3)-induced differentiation and accelerated the restoration of normal hematopoiesis. Theophylline, a well-tolerated phosphodiesterase inhibitor which stabilizes endogenous cAMP, also impaired APL growth and enhanced spontaneous or As(2)O(3)-triggered cell differentiation in vivo. Accordingly, in an APL patient resistant to combined RA-As(2)O(3) therapy, theophylline induced blast clearance and restored normal hematopoiesis. Taken together, these results demonstrate that in vivo activation of cAMP signaling contributes to APL clearance, independently of its RA-sensitivity, thus raising hopes that other myeloid leukemias may benefit from this therapeutic approach.
Collapse
Affiliation(s)
- Marie-Claude Guillemin
- CNRS UPR 9051, Laboratoire Associé au Comité de Paris de la Ligue contre le Cancer, affilié à l'Université de ParisVII, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lutz PG, Moog-Lutz C, Cayre YE. Signaling revisited in acute promyelocytic leukemia. Leukemia 2002; 16:1933-9. [PMID: 12357345 DOI: 10.1038/sj.leu.2402728] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2002] [Accepted: 07/03/2002] [Indexed: 11/09/2022]
Abstract
Although transcription factors are still the main focus to understanding leukemogenesis, recent results strongly suggest that alteration of a receptor and/or subsequent signaling plays a critical and co-operative role in the pathogenesis of acute myeloid leukemia (AML). The t(15;17) translocation, found in 95% of APL, encodes a PML-RARalpha fusion protein. A main model proposed for acute promyelocytic leukemia (APL) is that PML-RARalpha exerts its oncogenic effects by repressing retinoic acid-inducible genes critical to myeloid differentiation. Dysregulation of these genes may result in abnormal signaling, thereby freeing pre-leukemic cells from controls which normally induce the onset of differentiation. It is also likely that treatment of APL cells by retinoic acid induces de novo up-regulation of the same genes which are dominantly repressed by PML-RARalpha and whose expression is required for reactivation of the differentiation program. Identification of such genes together with the signaling pathways interrupted at the early stages of leukemia transformation and reactivated during retinoic acid-induced differentiation in APL cells will contribute to the development of new molecular targets for treatment of leukemia.
Collapse
MESH Headings
- Chromosomes, Human, Pair 15
- Chromosomes, Human, Pair 17
- Humans
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Signal Transduction
- Translocation, Genetic
Collapse
Affiliation(s)
- P G Lutz
- Unité INSERM U417, Hôpital Saint Antoine, Paris, France
| | | | | |
Collapse
|
48
|
Abstract
Despite its many therapeutic qualities, arsenic trioxide has been more commonly remembered as Madame Bovary's poison than as an anticancer drug. The ability of arsenic trioxide to treat acute promyelocytic leukaemia has radically changed this view, providing new insights into the pathogenesis of this malignancy and raising hopes that arsenicals might be useful in treating other cancers.
Collapse
MESH Headings
- Animals
- Arsenic Trioxide
- Arsenicals/history
- Arsenicals/therapeutic use
- Cell Differentiation
- China
- Europe
- Gene Expression Regulation, Neoplastic
- History, 15th Century
- History, 16th Century
- History, 18th Century
- History, 20th Century
- History, 21st Century
- History, Ancient
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Neoplasm Proteins/metabolism
- Nuclear Proteins
- Oxides/history
- Oxides/therapeutic use
- Promyelocytic Leukemia Protein
- Receptors, Retinoic Acid/metabolism
- Transcription Factors/metabolism
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- Jun Zhu
- CNRS UPR 9051, Laboratoire associé du comité de Paris de la ligue contre le cancer, affilié à l'université de Paris VII, Hôpital St Louis, 1 avenue C. Vellefaux, 75475 Paris cedex 10, France
| | | | | | | |
Collapse
|
49
|
Chen Z, Chen GQ, Shen ZX, Sun GL, Tong JH, Wang ZY, Chen SJ. Expanding the use of arsenic trioxide: leukemias and beyond. Semin Hematol 2002; 39:22-6. [PMID: 12012319 DOI: 10.1053/shem.2002.33611] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Arsenic has a long history of use in Chinese and Western medicine but fell out of use in the mid-20th century because of the unacceptable side effects that occurred at the doses that were thought to be necessary. The re-emergence of arsenic trioxide (ATO) in clinical use is due largely to purification of this compound from traditional mixtures, and the definition of effective, low-dose regimens for the treatment of acute promyelocytic leukemia (APL). ATO was first purified and used in controlled studies in patients with APL in China in the 1970s. Studies have subsequently also been performed in the United States. Complete response (CR) rates reported in patients with relapsed or refractory APL have varied from 52% to 92%, with similar rates reported in patients with newly diagnosed disease. The mechanism of action of ATO suggests it may be active against other malignancies, and ATO has shown some activity in patients with accelerated phase chronic myelogenous leukemia (CML) and multiple myeloma (MM). Clinical trials are ongoing and planned to define the optimal use of this compound in hematologic malignancies. Preliminary results from studies in patients with primary hepatocellular and gallbladder tumors indicate that ATO may also prove active against some solid tumors.
Collapse
Affiliation(s)
- Zhu Chen
- Institute of Hematology, Institutes of Clinical Research, Rui-Jin Hospital, Shanghai Second Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|