1
|
Liu J, Pan Y, Liu Y, Wei W, Hu X, Xin W, Chen N. The regulation of PTEN: Novel insights into functions as cancer biomarkers and therapeutic targets. J Cell Physiol 2023; 238:1693-1715. [PMID: 37334436 DOI: 10.1002/jcp.31053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023]
Abstract
This review summarizes the implications of the primary tumor suppressor protein phosphatase and tensin homolog (PTEN) in aggressive cancer development. PTEN interacts with other cellular proteins or factors suggesting the existence of an intricate molecular network that regulates their oncogenic function. Accumulating evidence has shown that PTEN exists and plays a role in the cytoplasmic organelles and in the nucleus. PTEN blocks phosphoinositide 3-kinases (PI3K)-protein kinase B-mammalian target of rapamycin signaling pathway by dephosphorylating phosphatidylinositol (PI)-3,4,5-triphosphate to PI-4,5-bisphosphate thus counteracting PI3K function. Studies have shown that PTEN expression is tightly regulated at transcriptional, posttranscriptional, and posttranslational levels (including protein-protein interactions and posttranslational modifications). Despite recent advances in PTEN research, the regulation and function of the PTEN gene remain largely unknown. How mutation or loss of specific exons in the PTEN gene occurs and involves in cancer development is not clear. This review illustrates the regulatory mechanisms of PTEN expression and discusses how PTEN participates in tumor development and/or suppression. Future prospects for the clinical applications are also highlighted.
Collapse
Affiliation(s)
- Jie Liu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yongli Pan
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Yuheng Liu
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaoping Hu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenqiang Xin
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Nan Chen
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
2
|
Jin N, Xia Y, Gao Q. Combined PARP inhibitors and small molecular inhibitors in solid tumor treatment (Review). Int J Oncol 2023; 62:28. [PMID: 36601757 PMCID: PMC9851129 DOI: 10.3892/ijo.2023.5476] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023] Open
Abstract
With the development of precision medicine, targeted therapy has attracted extensive attention. Poly(ADP‑ribose) polymerase inhibitors (PARPi) are critical clinical drugs designed to induce cell death and are major antitumor targeted agents. However, preclinical and clinical data have revealed the limitations of PARPi monotherapy. Therefore, their combination with other targeted drugs has become a research hotspot in tumor treatment. Recent studies have demonstrated the critical role of small molecular inhibitors in multiple haematological cancers and solid tumors via cellular signalling modulation, exhibiting potential as a combined pharmacotherapy. In the present review, studies focused on small molecular inhibitors targeting the homologous recombination pathway were summarized and clinical trials evaluating the safety and efficacy of combined treatment were discussed.
Collapse
Affiliation(s)
- Ning Jin
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Yu Xia
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Qinglei Gao
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
3
|
Zhao Z, Wang S, Zucknick M, Aittokallio T. Tissue-specific identification of multi-omics features for pan-cancer drug response prediction. iScience 2022; 25:104767. [PMID: 35992090 PMCID: PMC9385562 DOI: 10.1016/j.isci.2022.104767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Current statistical models for drug response prediction and biomarker identification fall short in leveraging the shared and unique information from various cancer tissues and multi-omics profiles. We developed mix-lasso model that introduces an additional sample group penalty term to capture tissue-specific effects of features on pan-cancer response prediction. The mix-lasso model takes into account both the similarity between drug responses (i.e., multi-task learning), and the heterogeneity between multi-omics data (multi-modal learning). When applied to large-scale pharmacogenomics dataset from Cancer Therapeutics Response Portal, mix-lasso enabled accurate drug response predictions and identification of tissue-specific predictive features in the presence of various degrees of missing data, drug-drug correlations, and high-dimensional and correlated genomic and molecular features that often hinder the use of statistical approaches in drug response modeling. Compared to tree lasso model, mix-lasso identified a smaller number of tissue-specific features, hence making the model more interpretable and stable for drug discovery applications. Pan-cancer cell lines provide a test bench for exploring gene-drug relationships Multi-omics data were integrated with pharmacological profiles for joint modeling Mix-lasso identifies tissue-specific biomarkers predictive of multi-drug responses Mix-lasso provides small number of stable features for drug discovery applications
Collapse
Affiliation(s)
- Zhi Zhao
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Norway
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Norway
| | - Shixiong Wang
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Norway
| | - Manuela Zucknick
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Norway
- Corresponding author
| | - Tero Aittokallio
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Norway
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Norway
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Finland
- Corresponding author
| |
Collapse
|
4
|
Heinemann L, Möllers KM, Ahmed HMM, Wei L, Sun K, Nimmagadda SC, Frank D, Baumann A, Poos AM, Dugas M, Varghese J, Raab MS, Khandanpour C. Inhibiting PI3K–AKT–mTOR Signaling in Multiple Myeloma-Associated Mesenchymal Stem Cells Impedes the Proliferation of Multiple Myeloma Cells. Front Oncol 2022; 12:874325. [PMID: 35795041 PMCID: PMC9251191 DOI: 10.3389/fonc.2022.874325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
The microenvironment of cancer cells is receiving increasing attention as an important factor influencing the progression and prognosis of tumor diseases. In multiple myeloma (MM), a hematological cancer of plasma cells, mesenchymal stem cells (MSCs) represent an integral part of the bone marrow niche and tumor microenvironment. It has been described that MM cells alter MSCs in a way that MM-associated MSCs promote the proliferation and survival of MM cells. Yet, our understanding of the molecular mechanisms governing the interaction between MM cells and MSCs and whether this can be targeted for therapeutic interventions is limited. To identify potential molecular targets, we examined MSCs by RNA sequencing and Western blot analysis. We report that MSCs from MM patients with active disease (MM-Act-MSCs) show a distinct gene expression profile as compared with MSCs from patients with other (non-) malignant diseases (CTR-MSCs). Of note, we detected a significant enrichment of the PI3K–AKT–mTOR hallmark gene set in MM-Act-MSCs and further confirmed the increased levels of related proteins in these MSCs. Pictilisib, a pan-PI3K inhibitor, selectively reduced the proliferation of MM-Act-MSCs as compared with CTR-MSCs. Furthermore, pictilisib treatment impaired the MM-promoting function of MM-Act-MSCs. Our data thus provide a deeper insight into the molecular signature and function of MSCs associated with MM and show that targeting PI3K–AKT–mTOR signaling in MSCs may represent an additional therapeutic pathway in the treatment of MM patients.
Collapse
Affiliation(s)
- Luca Heinemann
- Medical Department A, University Hospital Münster, Münster, Germany
| | | | | | - Lanying Wei
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Kaiyan Sun
- Medical Department A, University Hospital Münster, Münster, Germany
| | | | - Daria Frank
- Medical Department A, University Hospital Münster, Münster, Germany
| | - Anja Baumann
- Clinical Cooperation Unit (CCU) Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexandra M. Poos
- Clinical Cooperation Unit (CCU) Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Dugas
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Marc-Steffen Raab
- Clinical Cooperation Unit (CCU) Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Cyrus Khandanpour
- Medical Department A, University Hospital Münster, Münster, Germany
- Department of Haematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany
- *Correspondence: Cyrus Khandanpour,
| |
Collapse
|
5
|
Mezynski MJ, Farrelly AM, Cremona M, Carr A, Morgan C, Workman J, Armstrong P, McAuley J, Madden S, Fay J, Sheehan KM, Kay EW, Holohan C, Elamin Y, Rafee S, Morris PG, Breathnach O, Grogan L, Hennessy BT, Toomey S. Targeting the PI3K and MAPK pathways to improve response to HER2-targeted therapies in HER2-positive gastric cancer. J Transl Med 2021; 19:184. [PMID: 33933113 PMCID: PMC8088633 DOI: 10.1186/s12967-021-02842-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/18/2021] [Indexed: 12/24/2022] Open
Abstract
Background Aberrant PI3K signalling is implicated in trastuzumab resistance in HER2-positive gastric cancer (GC). The role of PI3K or MEK inhibitors in sensitising HER2-positive GCs to trastuzumab or in overcoming trastuzumab resistance is unclear. Methods Using mass spectrometry-based genotyping we analysed 105 hotspot, non-synonymous somatic mutations in PIK3CA and ERBB-family (EGFR, ERBB2, ERBB3 and ERBB4) genes in gastric tumour samples from 69 patients. A panel of gastric cell lines (N87, OE19, ESO26, SNU16, KATOIII) were profiled for anti-proliferative response to the PI3K inhibitor copanlisib and the MEK1/2 inhibitor refametinib alone and in combination with anti-HER2 therapies. Results Patients with HER2-positive GC had significantly poorer overall survival compared to HER2-negative patients (15.9 months vs. 35.7 months). Mutations in PIK3CA were only identified in HER2-negative tumours, while ERBB-family mutations were identified in HER2-positive and HER2-negative tumours. Copanlisib had anti-proliferative effects in 4/5 cell lines, with IC50s ranging from 23.4 (N87) to 93.8 nM (SNU16). All HER2-positive cell lines except SNU16 were sensitive to lapatinib (IC50s 0.04 µM–1.5 µM). OE19 cells were resistant to trastuzumab. The combination of lapatinib and copanlisib was synergistic in ESO-26 and OE-19 cells (ED50: 0.83 ± 0.19 and 0.88 ± 0.13, respectively) and additive in NCI-N87 cells (ED50:1.01 ± 0.55). The combination of copanlisib and trastuzumab significantly improved growth inhibition compared to either therapy alone in NCI-N87, ESO26 and OE19 cells (p < 0.05). Conclusions PI3K or MEK inhibition alone or in combination with anti-HER2 therapy may represent an improved treatment strategy for some patients with HER2-positive GC, and warrants further investigation in a clinical trial setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02842-1.
Collapse
Affiliation(s)
- M Janusz Mezynski
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Angela M Farrelly
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Mattia Cremona
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Aoife Carr
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Clare Morgan
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Julie Workman
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Paul Armstrong
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Jennifer McAuley
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Joanna Fay
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Katherine M Sheehan
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elaine W Kay
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ciara Holohan
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Yasir Elamin
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Shereen Rafee
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Patrick G Morris
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Oscar Breathnach
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Liam Grogan
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland.,Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
6
|
Krause G, Hassenrück F, Hallek M. Copanlisib for treatment of B-cell malignancies: the development of a PI3K inhibitor with considerable differences to idelalisib. Drug Des Devel Ther 2018; 12:2577-2590. [PMID: 30174412 PMCID: PMC6109662 DOI: 10.2147/dddt.s142406] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
On the occasion of its recent approval for relapsed follicular lymphoma, we review the design and development of the pan-class I PI3K inhibitor copanlisib as a drug for the treatment of B-cell malignancies in comparison with other kinase inhibitors targeting B-cell-receptor signaling, in particular with strictly isoform-δ-selective idelalisib. In agreement with previously defined PI3K-inhibitor chemotypes, the 2,3-dihydroimidazo[1,2-c]quinazoline scaffold of copanlisib adopts a flat conformation in the adenine-binding pocket of the catalytic p110 subunit and further extends into a deeper-affinity pocket in contrast to idelalisib, the quinazoline moiety of which is accommodated in a newly created selectivity pocket. Copanlisib shows higher potency than other clinically developed PI3K inhibitors against all four class I isoforms, with approximately tenfold preference for p110α and p110δ. Owing to its potency and isoform profile, copanlisib exhibits cell-type-specific cytotoxicity against primary chronic lymphocytic leukemia cells and diffuse large B-cell lymphoma (DLBCL) cell lines at nanomolar concentrations. Moreover, copanlisib differs from idelalisib in regard to intravenous versus oral administration and weekly versus twice-daily dosing. In regard to adverse effects, intermittent intravenous treatment with copanlisib leads to fewer gastrointestinal toxicities compared with continuous oral dosing of idelalisib. In relapsed follicular lymphoma, copanlisib appears more effective and especially better tolerated than other targeted therapies. Copanlisib extends existing treatment options for this subtype of indolent non-Hodgkin lymphoma and also shows promising response rates in DLBCL, especially of the activated B-cell type.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Purines/chemistry
- Purines/pharmacology
- Pyrimidines/chemistry
- Pyrimidines/pharmacology
- Quinazolines/chemistry
- Quinazolines/pharmacology
- Quinazolinones/chemistry
- Quinazolinones/pharmacology
Collapse
Affiliation(s)
- Günter Krause
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Köln Bonn, Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany,
| | - Floyd Hassenrück
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Köln Bonn, Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany,
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Köln Bonn, Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany,
| |
Collapse
|
7
|
Cross Talk Networks of Mammalian Target of Rapamycin Signaling With the Ubiquitin Proteasome System and Their Clinical Implications in Multiple Myeloma. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 343:219-297. [PMID: 30712673 DOI: 10.1016/bs.ircmb.2018.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy and results from the clonal amplification of plasma cells. Despite recent advances in treatment, MM remains incurable with a median survival time of only 5-6years, thus necessitating further insights into MM biology and exploitation of novel therapeutic approaches. Both the ubiquitin proteasome system (UPS) and the PI3K/Akt/mTOR signaling pathways have been implicated in the pathogenesis, and treatment of MM and different lines of evidence suggest a close cross talk between these central cell-regulatory signaling networks. In this review, we outline the interplay between the UPS and mTOR pathways and discuss their implications for the pathophysiology and therapy of MM.
Collapse
|
8
|
Abstract
Bayer are developing copanlisib (Aliqopa™)-a pan-class I phosphoinositide 3-kinase (PI3K) inhibitor-as a treatment for various haematological and solid malignancies. The US FDA has granted copanlisib accelerated approval for the treatment of adults with relapsed follicular lymphoma who have received at least two prior systemic therapies based on the results of a phase II trial. Phase III trials are underway evaluating copanlisib as treatment for relapsed/refractory diffuse large B-cell lymphoma and in combination with rituximab or rituximab-based chemotherapy or standard immunochemotherapy in patients with relapsed indolent B-cell non-Hodgkin's lymphoma. Phase I/II studies are underway in relapsed or refractory peripheral T-cell or NK/T-cell lymphoma, advanced cholangiocarcinoma, hormone receptor-positive HER2-negative stage I-IV breast cancer, HER2-positive breast cancer and recurrent and/or metastatic head and neck squamous cell carcinomas harbouring a PI3KCA mutation/amplification and/or a PTEN loss. This article summarizes the milestones in the development of copanlisib leading to this first approval for relapsed follicular lymphoma.
Collapse
Affiliation(s)
- Anthony Markham
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand.
| |
Collapse
|
9
|
Abramson HN. Kinase inhibitors as potential agents in the treatment of multiple myeloma. Oncotarget 2018; 7:81926-81968. [PMID: 27655636 PMCID: PMC5348443 DOI: 10.18632/oncotarget.10745] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
Recent years have witnessed a dramatic increase in the number of therapeutic options available for the treatment of multiple myeloma (MM) - from immunomodulating agents to proteasome inhibitors to histone deacetylase (HDAC) inhibitors and, most recently, monoclonal antibodies. Used in conjunction with autologous hematopoietic stem cell transplantation, these modalities have nearly doubled the disease's five-year survival rate over the last three decades to about 50%. In spite of these advances, MM still is considered incurable as resistance and relapse are common. While small molecule protein kinase inhibitors have made inroads in the therapy of a number of cancers, to date their application to MM has been less than successful. Focusing on MM, this review examines the roles played by a number of kinases in driving the malignant state and the rationale for target development in the design of a number of kinase inhibitors that have demonstrated anti-myeloma activity in both in vitro and in vivo xenograph models, as well as those that have entered clinical trials. Among the targets and their inhibitors examined are receptor and non-receptor tyrosine kinases, cell cycle control kinases, the PI3K/AKT/mTOR pathway kinases, protein kinase C, mitogen-activated protein kinase, glycogen synthase kinase, casein kinase, integrin-linked kinase, sphingosine kinase, and kinases involved in the unfolded protein response.
Collapse
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
10
|
Patnaik A, Appleman LJ, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, Weiss GJ, Sachdev JC, Chadha M, Fulk M, Ejadi S, Mountz JM, Lotze MT, Toledo FGS, Chu E, Jeffers M, Peña C, Xia C, Reif S, Genvresse I, Ramanathan RK. First-in-human phase I study of copanlisib (BAY 80-6946), an intravenous pan-class I phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors and non-Hodgkin's lymphomas. Ann Oncol 2017; 27:1928-40. [PMID: 27672108 PMCID: PMC5035790 DOI: 10.1093/annonc/mdw282] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND To evaluate the safety, tolerability, pharmacokinetics, and maximum tolerated dose (MTD) of copanlisib, a phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors or non-Hodgkin's lymphoma (NHL). PATIENTS AND METHODS Phase I dose-escalation study including patients with advanced solid tumors or NHL, and a cohort of patients with type 2 diabetes mellitus. Patients received three weekly intravenous infusions of copanlisib per 28-day cycle over the dose range 0.1-1.2 mg/kg. Plasma copanlisib levels were analyzed for pharmacokinetics. Biomarker analysis included PIK3CA, KRAS, BRAF, and PTEN mutational status and PTEN immunohistochemistry. Whole-body [(18)F]-fluorodeoxyglucose positron emission tomography ((18)FDG-PET) was carried out at baseline and following the first dose to assess early pharmacodynamic effects. Plasma glucose and insulin levels were evaluated serially. RESULTS Fifty-seven patients received treatment. The MTD was 0.8 mg/kg copanlisib. The most frequent treatment-related adverse events were nausea and transient hyperglycemia. Copanlisib exposure was dose-proportional with no accumulation; peak exposure positively correlated with transient hyperglycemia post-infusion. Sixteen of 20 patients treated at the MTD had reduced (18)FDG-PET uptake; 7 (33%) had a reduction >25%. One patient achieved a complete response (CR; endometrial carcinoma exhibiting both PIK3CA and PTEN mutations and complete PTEN loss) and two had a partial response (PR; both metastatic breast cancer). Among the nine NHL patients, all six with follicular lymphoma (FL) responded (one CR and five PRs) and one patient with diffuse large B-cell lymphoma had a PR by investigator assessment; two patients with FL who achieved CR (per post hoc independent radiologic review) were on treatment >3 years. CONCLUSION Copanlisib, dosed intermittently on days 1, 8, and 15 of a 28-day cycle, was well tolerated and the MTD was determined to be 0.8 mg/kg. Copanlisib exhibited dose-proportional pharmacokinetics and promising anti-tumor activity, particularly in patients with NHL. CLINICALTRIALSGOV NCT00962611; https://clinicaltrials.gov/ct2/show/NCT00962611.
Collapse
Affiliation(s)
- A Patnaik
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | | | - A W Tolcher
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - K P Papadopoulos
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - M Beeram
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - D W Rasco
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - G J Weiss
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale Cancer Treatment Centers of America, Goodyear
| | - J C Sachdev
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | - M Chadha
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | - M Fulk
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | - S Ejadi
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | | | - M T Lotze
- University of Pittsburgh, Pittsburgh
| | | | - E Chu
- University of Pittsburgh, Pittsburgh
| | - M Jeffers
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, USA
| | - C Peña
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, USA
| | - C Xia
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, USA
| | - S Reif
- Bayer Pharma AG, Berlin, Germany
| | | | - R K Ramanathan
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| |
Collapse
|
11
|
Yu Y, Hall T, Eathiraj S, Wick MJ, Schwartz B, Abbadessa G. In-vitro and in-vivo combined effect of ARQ 092, an AKT inhibitor, with ARQ 087, a FGFR inhibitor. Anticancer Drugs 2017; 28:503-513. [PMID: 28240679 PMCID: PMC5404396 DOI: 10.1097/cad.0000000000000486] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The PI3K/AKT pathway plays an important role in the initiation and progression of cancer, and the drug development efforts targeting this pathway with therapeutic interventions have been advanced by academic and industrial groups. However, the clinical outcome is moderate. Combination of inhibition of PI3K/AKT and other targeted agents became a feasible approach. In this study we assessed the combined effect of ARQ 092, a pan-AKT inhibitor, and ARQ 087, a pan-FGFR inhibitor, in vitro and in vivo. In a panel of 45 cancer cell lines, on 24% (11 out of 45) the compounds showed synergistic effect, on 62% (28 out of 45) additive, and on 13% (6 out of 45) antagonistic. The highest percentage of synergism was found on endometrial and ovarian cancer cell lines. Mutational analysis revealed that PIK3CA/PIK3R1 mutations and aberrant activation of FGFR2 predicted synergism, whereas Ras mutations showed a reverse correlation. Pathway analysis revealed that a combination of ARQ 092 and ARQ 087 enhanced the inhibition of both the AKT and FGFR pathways in cell lines in which synergistic effects were found (AN3CA and IGROV-1). Cell cycle arrest and apoptotic response occurred only in AN3CA cell, and was not seen in IGROV-1 cells. Furthermore, enhanced antitumor activity was observed in mouse models with endometrial cancer cell line and patient-derived tumors when ARQ 092 and ARQ 087 were combined. These results from in-vitro and in-vivo studies provide a strong rationale in treating endometrial and other cancers with the activated PI3K/AKT and FGFR pathways.
Collapse
Affiliation(s)
- Yi Yu
- aArQule, Inc., Burlington, Massachusetts bSouth Texas Accelerated Research Therapeutics, San Antonio, Texas, USA
| | | | | | | | | | | |
Collapse
|
12
|
Gavriatopoulou M, Dimopoulos MA, Kastritis E, Terpos E. Emerging treatment approaches for myeloma-related bone disease. Expert Rev Hematol 2017; 10:217-228. [PMID: 28092987 DOI: 10.1080/17474086.2017.1283213] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Multiple myeloma is characterized by the presence of osteolytic lesions that leads to devastating skeletal-related events in the majority of patients. Myeloma bone disease is attributed to increased osteoclastic and suppressed osteoblastic activity. Areas covered: Bisphosphonates remain the main treatment option, however they have limitations on their own. Understanding the pathogenesis of myeloma bone disease may provide a roadmap for new therapeutic approaches. The pathway of RANKRANKLOPG pathway has revealed denosumab, a monoclonal antibody targeting RANKL as a novel emerging therapy for myeloma-related bone disease. Furthermore, the Wnt signaling inhibitors dicckopf-1 and sclerostin that are implicated in the pathogenesis of bone destruction of myeloma are now targeted by novel monoclonal antibodies. Activin-A is a TGF-beta superfamily member which increases osteoclast activity and inhibits osteoblast function in myeloma; sotatercept and other molecules targeting activin-A have entered into clinical development. Several other molecules and pathways that play an important role in the pathogenesis of bone destruction in myeloma, such as periostin, adiponectin, Notch and BTK signaling are also targeted in an attempt to develop novel therapies for myeloma-related bone disease. Expert commentary: We summarize the current advances in the biology of myeloma bone disease and the potential therapeutic targets.
Collapse
Affiliation(s)
- Maria Gavriatopoulou
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| | - Meletios A Dimopoulos
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| | - Efstathios Kastritis
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| | - Evangelos Terpos
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| |
Collapse
|
13
|
Xi H, An R, Li L, Wang G, Tao Y, Gao L. Myeloma bone disease: Progress in pathogenesis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:149-155. [PMID: 27496181 DOI: 10.1016/j.pbiomolbio.2016.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/31/2016] [Accepted: 08/02/2016] [Indexed: 12/12/2022]
Abstract
Myeloma bone disease (MBD) is one of the most serious complications of multiple myeloma (MM) and the most severe cause of MM morbidity. Dysregulation of osteoblast and osteoclast cells plays key roles in MBD. In the bone marrow microenvironment, myeloma cells, osteoblasts, osteoclasts and bone marrow stromal cells can secrete multiple cytokines, categorized as osteoclast cell activating factors (OAFs) and osteoblast cell inactivating factors, which have been discovered to participate in bone metabolism and contribute to the pathogenesis of MBD. Several signaling pathways related to these cytokines were also revealed in the MBD pathogenesis. To better understand the pathogenesis of MBD and therefore the potential therapeutic targets of this disease, we will summarize recent study progress in the factors and underlying signaling pathways involved in the occurrence and development of MBD.
Collapse
Affiliation(s)
- Hao Xi
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ran An
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Lu Li
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Gang Wang
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yi Tao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Lu Gao
- Department of Physiology, Second Military Medical University, Shanghai, China.
| |
Collapse
|
14
|
Yu W, Chen Y, Xiang R, Xu W, Wang Y, Tong J, Zhang N, Wu Y, Yan H. Novel phosphatidylinositol 3-kinase inhibitor BKM120 enhances the sensitivity of multiple myeloma to bortezomib and overcomes resistance. Leuk Lymphoma 2016; 58:428-437. [PMID: 27439454 DOI: 10.1080/10428194.2016.1190968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Proteasome inhibitor bortezomib has proven efficacy against multiple myeloma. However, bortezomib activates the phosphatidylinositol 3-kinase/AKT (PI3K/AKT) pathway (which is essential to the development of myeloma), often resulting in drug resistance and disease recurrence. The addition of BKM120 significantly enhanced the apoptotic effects of bortezomib in both bortezomib-sensitive and bortezomib-resistant cells. Treatment with bortezomib alone increased the phosphorylation of AKT (P-AKT), whereas the addition of BKM120 markedly downregulated P-AKT in both bortezomib-sensitive and bortezomib-resistant cells. The clinical relevance of combined treatment with bortezomib and BKM120 was investigated in a xenograft mouse model and in myeloma patients, and the synergy of the combination was confirmed. In conclusion, the addition of BKM120 enhanced the sensitivity of myeloma cells to bortezomib.
Collapse
Affiliation(s)
- Wenjun Yu
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Yubao Chen
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Rufang Xiang
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Wenbin Xu
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Yan Wang
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Jia Tong
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Nan Zhang
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Yingli Wu
- b Hongqiao International Institute of Medicine, Shanghai Tongren Hospital , Shanghai , China.,c Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education , School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Hua Yan
- a Department of Hematology , Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| |
Collapse
|
15
|
Naymagon L, Abdul-Hay M. Novel agents in the treatment of multiple myeloma: a review about the future. J Hematol Oncol 2016; 9:52. [PMID: 27363832 PMCID: PMC4929712 DOI: 10.1186/s13045-016-0282-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023] Open
Abstract
Multiple myeloma (MM) is a disease that affects plasma cells and can lead to devastating clinical features such as anemia, lytic bone lesions, hypercalcemia, and renal disease. An enhanced understanding of MM disease mechanisms has led to new more targeted treatments. There is now a plethora of treatments available for MM. In this review article, our aim is to discuss many of the novel agents that are being studied or have recently been approved for the treatment of MM. These agents include the following: immunomodulators (pomalidomide), proteasome inhibitors (carfilzomib, marizomib, ixazomib, oprozomib), alkylating agents (bendamustine), AKT inhibitors (afuresertib), BTK inhibitors (ibrutinib), CDK inhibitors (dinaciclib), histone deacetylase inhibitors (panobinostat, rocilinostat, vorinostat), IL-6 inhibitors (siltuximab), kinesin spindle protein inhibitors (filanesib), monoclonal antibodies (daratumumab, elotuzumab, indatuximab, SAR650984), and phosphoinositide 3-kinase (PI3K) inhibitors.
Collapse
Affiliation(s)
| | - Maher Abdul-Hay
- Department of Medicine, New York University, New York, USA. .,Perlmutter Cancer Center, New York University, New York, USA. .,NYU School of Medicine, 240 East 38th Street, 19 Floor, New York, NY, 10016, USA.
| |
Collapse
|
16
|
Schneider P, Schön M, Pletz N, Seitz CS, Liu N, Ziegelbauer K, Zachmann K, Emmert S, Schön MP. The novel PI3 kinase inhibitor, BAY 80-6946, impairs melanoma growth in vivo and in vitro. Exp Dermatol 2016; 23:579-84. [PMID: 24942196 DOI: 10.1111/exd.12470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2014] [Indexed: 12/14/2022]
Abstract
Due to its almost universal resistance to chemotherapy, metastasized melanoma remains a major challenge in clinical oncology. Given that phosphatidyl inositol-3 kinase (PI3K) activation in melanoma cells is associated with poor prognosis, disease progression and resistance to chemotherapy, the PI3K-Akt signalling pathway is a promising therapeutic target for melanoma treatment. We analysed six human melanoma cell lines for their constitutive activation of Akt and then tested two representative lines, A375 and LOX, for their susceptibility to PI3K-inhibition by the highly specific small molecule inhibitor, BAY 80-6946. In addition, the effect of BAY 80-6946 on A375 and LOX melanoma cells was assessed in vivo in a xenotransplantation mouse model. We provide experimental evidence that specifically inhibiting the PI3K pathway and phosphorylation of Akt by this novel compound results in antitumoral activities including inhibition of proliferation, induction of apoptosis and cell cycle arrest in vitro and in vivo. However, the susceptibility did not show a clear-cut pattern and differed between the melanoma cell lines tested, resulting in in vivo growth inhibition of A375 but not LOX melanoma cells. Thus, in some cases BAY 80-6946 or related compounds may be a valuable addition to the therapeutic armamentarium.
Collapse
Affiliation(s)
- Philine Schneider
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Systemic modeling myeloma-osteoclast interactions under normoxic/hypoxic condition using a novel computational approach. Sci Rep 2015; 5:13291. [PMID: 26282073 PMCID: PMC4539608 DOI: 10.1038/srep13291] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/20/2015] [Indexed: 12/17/2022] Open
Abstract
Interaction of myeloma cells with osteoclasts (OC) can enhance tumor cell expansion through activation of complex signaling transduction networks. Both cells reside in the bone marrow, a hypoxic niche. How OC-myeloma interaction in a hypoxic environment affects myeloma cell growth and their response to drug treatment is poorly understood. In this study, we i) cultured myeloma cells in the presence/absence of OCs under normoxia and hypoxia conditions and did protein profiling analysis using reverse phase protein array; ii) computationally developed an Integer Linear Programming approach to infer OC-mediated myeloma cell-specific signaling pathways under normoxic and hypoxic conditions. Our modeling analysis indicated that in the presence OCs, (1) cell growth-associated signaling pathways, PI3K/AKT and MEK/ERK, were activated and apoptotic regulatory proteins, BAX and BIM, down-regulated under normoxic condition; (2) β1 Integrin/FAK signaling pathway was activated in myeloma cells under hypoxic condition. Simulation of drug treatment effects by perturbing the inferred cell-specific pathways showed that targeting myeloma cells with the combination of PI3K and integrin inhibitors potentially (1) inhibited cell proliferation by reducing the expression/activation of NF-κB, S6, c-Myc, and c-Jun under normoxic condition; (2) blocked myeloma cell migration and invasion by reducing the expression of FAK and PKC under hypoxic condition.
Collapse
|
18
|
Göckeritz E, Kerwien S, Baumann M, Wigger M, Vondey V, Neumann L, Landwehr T, Wendtner CM, Klein C, Liu N, Hallek M, Frenzel LP, Krause G. Efficacy of phosphatidylinositol-3 kinase inhibitors with diverse isoform selectivity profiles for inhibiting the survival of chronic lymphocytic leukemia cells. Int J Cancer 2015; 137:2234-42. [DOI: 10.1002/ijc.29579] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 03/13/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Elisa Göckeritz
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Susan Kerwien
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Michael Baumann
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Marion Wigger
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Verena Vondey
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Lars Neumann
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Thomas Landwehr
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Clemens M. Wendtner
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Christian Klein
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich; Switzerland
| | - Ningshu Liu
- Bayer HealthCare Pharmaceuticals, Global Drug Discovery, TRG Oncology; Berlin Germany
| | - Michael Hallek
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Lukas P. Frenzel
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Günter Krause
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| |
Collapse
|
19
|
Tang J, Zhu J, Yu Y, Zhang Z, Chen G, Zhou X, Qiao C, Hou T, Mao X. A virtual screen identified C96 as a novel inhibitor of phosphatidylinositol 3-kinase that displays potent preclinical activity against multiple myeloma in vitro and in vivo. Oncotarget 2015; 5:3836-48. [PMID: 25003534 PMCID: PMC4116524 DOI: 10.18632/oncotarget.1657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway is emerging as a promising therapeutic target for multiple myeloma (MM). In the present study, we performed a virtual screen against 800,000 of small molecule compounds by targeting PI3Kγ. C96, one of such compounds, inhibited PI3K activated by insulin-like growth factor-1 (IGF-1), but did not suppress IGF-1R activation. The cell-free assay revealed that C96 preferred to inhibit PI3Kα and δ, but was not active against AKT1, 2, 3 or mTOR. C96 inhibited PI3K activation in a time- and concentration-dependent manner. Consistent with its inhibition on PI3K/AKT, C96 downregulated the activation of mTOR, p70S6K, 4E-BP1, but did not suppress other kinases such as ERK and c-Src. Inhibition of the PI3K/AKT signaling pathway by C96 led to MM cell apoptosis which was demonstrated by Annexin V staining and activation of the pro-apoptotic signals. Furthermore, C96 displayed potent anti-myeloma activity in a MM xenograft model in nude mice. Oral administration of 100 mg/kg bodyweight almost fully suppressed tumor growth within 16 days, but without gross toxicity. Importantly, AKT activation was suppressed in tumor tissues from C96-treated mice, which was consistent with delayed tumor growth. Thus, we identified a novel PI3K inhibitor with a great potential for MM therapy.
Collapse
Affiliation(s)
- Juan Tang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China;Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China
| | | | | | | | | | | | | | | | - Xinliang Mao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China;Collaborative Innovation Center of Hematology, Suzhou, China
| |
Collapse
|
20
|
Schwartz S, Wongvipat J, Trigwell CB, Hancox U, Carver BS, Rodrik-Outmezguine V, Will M, Yellen P, de Stanchina E, Baselga J, Scher HI, Barry ST, Sawyers CL, Chandarlapaty S, Rosen N. Feedback suppression of PI3Kα signaling in PTEN-mutated tumors is relieved by selective inhibition of PI3Kβ. Cancer Cell 2015; 27:109-22. [PMID: 25544636 PMCID: PMC4293347 DOI: 10.1016/j.ccell.2014.11.008] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 09/25/2014] [Accepted: 11/08/2014] [Indexed: 01/16/2023]
Abstract
In PTEN-mutated tumors, we show that PI3Kα activity is suppressed and PI3K signaling is driven by PI3Kβ. A selective inhibitor of PI3Kβ inhibits the Akt/mTOR pathway in these tumors but not in those driven by receptor tyrosine kinases. However, inhibition of PI3Kβ only transiently inhibits Akt/mTOR signaling because it relieves feedback inhibition of IGF1R and other receptors and thus causes activation of PI3Kα and a rebound in downstream signaling. This rebound is suppressed and tumor growth inhibition enhanced with combined inhibition of PI3Kα and PI3Kβ. In PTEN-deficient models of prostate cancer, this effective inhibition of PI3K causes marked activation of androgen receptor activity. Combined inhibition of both PI3K isoforms and androgen receptor results in major tumor regressions.
Collapse
Affiliation(s)
- Sarit Schwartz
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - John Wongvipat
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Cath B Trigwell
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - Urs Hancox
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - Brett S Carver
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vanessa Rodrik-Outmezguine
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marie Will
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paige Yellen
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elisa de Stanchina
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - José Baselga
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Simon T Barry
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - Charles L Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Neal Rosen
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
21
|
Elster N, Cremona M, Morgan C, Toomey S, Carr A, O’Grady A, Hennessy BT, Eustace AJ. A preclinical evaluation of the PI3K alpha/delta dominant inhibitor BAY 80-6946 in HER2-positive breast cancer models with acquired resistance to the HER2-targeted therapies trastuzumab and lapatinib. Breast Cancer Res Treat 2014; 149:373-83. [DOI: 10.1007/s10549-014-3239-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 12/10/2014] [Indexed: 01/08/2023]
|
22
|
Distinct roles of class I PI3K isoforms in multiple myeloma cell survival and dissemination. Blood Cancer J 2014; 4:e204. [PMID: 24769645 PMCID: PMC4003418 DOI: 10.1038/bcj.2014.24] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
23
|
Dillon LM, Miller TW. Therapeutic targeting of cancers with loss of PTEN function. Curr Drug Targets 2014; 15:65-79. [PMID: 24387334 PMCID: PMC4310752 DOI: 10.2174/1389450114666140106100909] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/30/2013] [Accepted: 11/02/2013] [Indexed: 02/08/2023]
Abstract
Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is one of the most frequently disrupted tumor suppressors in cancer. The lipid phosphatase activity of PTEN antagonizes the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathway to repress tumor cell growth and survival. In the nucleus, PTEN promotes chromosome stability and DNA repair. Consequently, loss of PTEN function increases genomic instability. PTEN deficiency is caused by inherited germline mutations, somatic mutations, epigenetic and transcriptional silencing, post-translational modifications, and protein-protein interactions. Given the high frequency of PTEN deficiency across cancer subtypes, therapeutic approaches that exploit PTEN loss-of-function could provide effective treatment strategies. Herein, we discuss therapeutic strategies aimed at cancers with loss of PTEN function, and the challenges involved in treating patients afflicted with such cancers. We review preclinical and clinical findings, and highlight novel strategies under development to target PTENdeficient cancers.
Collapse
Affiliation(s)
| | - Todd W Miller
- Dartmouth-Hitchcock Medical Center, One Medical Center Dr. HB-7936, Lebanon, NH 03756, USA.
| |
Collapse
|