1
|
Lohrberg M, Heber M, Ries L, Markus K, Ksionsko N, Schmidt N, Nothnick G, Thielking L, O'Neill M, Martínéz-González S, Blanco-Aparicio C, Pastor J, Cunningham D, Koch R. Dual Targeting of Pim and PI3 Kinases in Mature T-Cell Lymphoma. Eur J Haematol 2025. [PMID: 40165380 DOI: 10.1111/ejh.14420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Provirus Integration site for Moloney leukemia virus (Pim) family members are well-known oncogenes, with an expression that is restricted to few cell types including hematopoietic cells in adult organisms, making it a promising target for lymphoma treatment. Indeed, previous studies in mature T-cell lymphoma (mTCL) cells revealed frequent upregulation of Pim expression. Nevertheless, inhibition of Pim kinases showed only minor effects on the viability of mTCL cells so far. Thus, we here addressed cellular responses to therapeutic inhibition of Pim kinases and identified a PI3K/Akt-driven activation of mTOR as a significant escape mechanism mitigating the anti-lymphoma effects of Pim inhibition. Indeed, dual inhibition of Pim and PI3 kinases showed synergistic anti-lymphoma effects in vitro through downregulation of mTOR-induced protein translation and mitigation of BCL-xL-mediated anti-apoptotic mechanisms. Based on this finding, we next explored the therapeutic potential of the dual Pim/PI3K inhibitor IBL-202 in mTCL cell lines. Strikingly, IBL-202 strongly induced cell-cycle-dependent cell death in cell lines of different mTCL subtypes. Together, our study provides mechanistic evidence supporting a therapeutic strategy of dual Pim- and PI3-kinase inhibition in mature T-cell lymphoma.
Collapse
Affiliation(s)
- M Lohrberg
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - M Heber
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - L Ries
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - K Markus
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - N Ksionsko
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - N Schmidt
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - G Nothnick
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - L Thielking
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - M O'Neill
- Inflection Biosciences Ltd., Dublin, Ireland
| | | | | | - J Pastor
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | | | - R Koch
- Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
2
|
PIM Kinases in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13174304. [PMID: 34503111 PMCID: PMC8428354 DOI: 10.3390/cancers13174304] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable disease and novel therapeutic agents/approaches are urgently needed. The PIM (Proviral insertion in murine malignancies) serine/threonine kinases have 3 isoforms: PIM1, PIM2, and PIM3. PIM kinases are engaged with an expansive scope of biological activities including cell growth, apoptosis, drug resistance, and immune response. An assortment of molecules and pathways that are critical to myeloma tumorigenesis has been recognized as the downstream targets of PIM kinases. The inhibition of PIM kinases has become an emerging scientific interest for the treatment of multiple myeloma and several PIM kinase inhibitors, such as SGI-1776, AZD1208, and PIM447 (formerly LGH447), have been developed and are under different phases of clinical trials. Current research has been focused on the development of a new generation of potent PIM kinase inhibitors with appropriate pharmacological profiles reasonable for human malignancy treatment. Combination therapy of PIM kinase inhibitors with chemotherapeutic appears to create an additive cytotoxic impact in cancer cells. Notwithstanding, the mechanisms by which PIM kinases modulate the immune microenvironment and synergize with the immunomodulatory agents such as lenalidomide have not been deliberately depicted. This review provides a comprehensive overview of the PIM kinase pathways and the current research status of the development of PIM kinase inhibitors for the treatment of MM. Additionally, the combinatorial effects of the PIM kinase inhibitors with other targeted agents and the promising strategies to exploit PIM as a therapeutic target in malignancy are highlighted.
Collapse
|
3
|
Mologni L, Marzaro G, Redaelli S, Zambon A. Dual Kinase Targeting in Leukemia. Cancers (Basel) 2021; 13:E119. [PMID: 33401428 PMCID: PMC7796318 DOI: 10.3390/cancers13010119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Pharmacological cancer therapy is often based on the concurrent inhibition of different survival pathways to improve treatment outcomes and to reduce the risk of relapses. While this strategy is traditionally pursued only through the co-administration of several drugs, the recent development of multi-targeting drugs (i.e., compounds intrinsically able to simultaneously target several macromolecules involved in cancer onset) has had a dramatic impact on cancer treatment. This review focuses on the most recent developments in dual-kinase inhibitors used in acute myeloid leukemia (AML), chronic myelogenous leukemia (CML), and lymphoid tumors, giving details on preclinical studies as well as ongoing clinical trials. A brief overview of dual-targeting inhibitors (kinase/histone deacetylase (HDAC) and kinase/tubulin polymerization inhibitors) applied to leukemia is also given. Finally, the very recently developed Proteolysis Targeting Chimeras (PROTAC)-based kinase inhibitors are presented.
Collapse
Affiliation(s)
- Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.M.); (S.R.)
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, I-35131 Padova, Italy;
| | - Sara Redaelli
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.M.); (S.R.)
| | - Alfonso Zambon
- Department of Chemistry and Geological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
4
|
Toth RK, Warfel NA. Targeting PIM Kinases to Overcome Therapeutic Resistance in Cancer. Mol Cancer Ther 2020; 20:3-10. [PMID: 33303645 DOI: 10.1158/1535-7163.mct-20-0535] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/24/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022]
Abstract
Cancer progression and the onset of therapeutic resistance are often the results of uncontrolled activation of survival kinases. The proviral integration for the Moloney murine leukemia virus (PIM) kinases are oncogenic serine/threonine kinases that regulate tumorigenesis by phosphorylating a wide range of substrates that control cellular metabolism, proliferation, and survival. Because of their broad impact on cellular processes that facilitate progression and metastasis in many cancer types, it has become clear that the activation of PIM kinases is a significant driver of resistance to various types of anticancer therapies. As a result, efforts to target PIM kinases for anticancer therapy have intensified in recent years. Clinical and preclinical studies indicate that pharmacologic inhibition of PIM has the potential to significantly improve the efficacy of standard and targeted therapies. This review focuses on the signaling pathways through which PIM kinases promote cancer progression and resistance to therapy, as well as highlights biological contexts and promising strategies to exploit PIM as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Rachel K Toth
- University of Arizona Cancer Center, Tucson, Arizona
| | - Noel A Warfel
- University of Arizona Cancer Center, Tucson, Arizona. .,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
5
|
IBL-202 is synergistic with venetoclax in CLL under in vitro conditions that mimic the tumor microenvironment. Blood Adv 2020; 4:5093-5106. [PMID: 33085757 DOI: 10.1182/bloodadvances.2019001369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 09/06/2020] [Indexed: 10/23/2022] Open
Abstract
The B-cell receptor signaling pathway and dysregulation of the Bcl-2 family of proteins play crucial roles in the pathogenesis of chronic lymphocytic leukemia (CLL). Despite significant advances in the treatment of the disease, relapse and drug resistance are not uncommon. In the current study, we investigated the dual PI3/PIM kinase inhibitor IBL-202 in combination with venetoclax as a treatment option for CLL using both primary CLL cells and TP53-deficient OSU-CLL cells generated using the CRISPR-Cas9 system. IBL-202 and venetoclax were highly synergistic against primary CLL cells cocultured with CD40L fibroblasts (combination index [CI], 0.4, at a fractional effect of 0.9) and TP53-knockout (KO) OSU-CLL cells (CI, 0.5, at a fractional effect of 0.9). Synergy between the drugs was consistent, with a significant (P < .05) reduction in the 50% inhibitory concentration for both drugs. IBL-202 and venetoclax in combination induced cell-cycle arrest and slowed the proliferation of both wild-type and TP53-KO cell lines. The drug combination inhibited AKT phosphorylation, reduced expression of Bcl-xL and NF-κB, and increased the Noxa/Mcl-1 ratio. Downregulation of CXCR4 was consistent with inhibition of the SDF-1α-induced migratory capacity of CLL cells. Synergy between IBL-202 and venetoclax against primary CLL cells cultured under conditions that mimic the tumor microenvironment suggests this drug combination may be effective against CLL cells within the lymph nodes and bone marrow. Furthermore, the efficacy of the combination against the TP53-KO OSU-CLL cell line suggests the combination may be a highly effective treatment strategy for high-risk CLL.
Collapse
|
6
|
Leung MS, Chan KKS, Dai WJ, Wong CY, Au KY, Wong PY, Wong CCL, Lee TKW, Ng IOL, Kao WJ, Lo RCL. Anti-tumour effects of PIM kinase inhibition on progression and chemoresistance of hepatocellular carcinoma. J Pathol 2020; 252:65-76. [PMID: 32558942 DOI: 10.1002/path.5492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is a biologically aggressive cancer. Targeted therapy is in need to tackle challenges in the treatment perspective. A growing body of evidence suggests a promising role of pharmacological inhibition of PIM (proviral integration site for Moloney murine leukaemia virus) kinase in some human haematological and solid cancers. Yet to date, the potential application of PIM inhibitors in HCC is still largely unexplored. In the present study we investigated the pre-clinical efficacy of PIM inhibition as a therapeutic approach in HCC. Effects of PIM inhibitors on cell proliferation, migration, invasion, chemosensitivity, and self-renewal were examined in vitro. The effects of PIM inhibitors on tumour growth and chemoresistance in vivo were studied using xenograft mouse models. Potential downstream molecular mechanisms were elucidated by RNA sequencing (RNA-seq) of tumour tissues harvested from animal models. Our findings demonstrate that PIM inhibitors SGI-1776 and PIM447 reduced HCC proliferation, metastatic potential, and self-renewal in vitro. Results from in vivo experiments supported the role of PIM inhibition in suppressing of tumour growth and increasing chemosensitivity of HCC toward cisplatin and doxorubicin, the two commonly used chemotherapeutic agents in trans-arterial chemoembolisation (TACE) for HCC. RNA-seq analysis revealed downregulation of the MAPK/ERK pathway upon PIM inhibition in HCC cells. In addition, LOXL2 and ICAM1 were identified as potential downstream effectors. Taken together, PIM inhibitors demonstrated remarkable anti-tumourigenic effects in HCC in vitro and in vivo. PIM kinase inhibition is a potential approach to be exploited in formulating adjuvant therapy for HCC patients of different disease stages. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | | | - Wen-Juan Dai
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Cheuk-Yan Wong
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Kwan-Yung Au
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Pik-Ying Wong
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Carmen Chak-Lui Wong
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, PR China.,State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong SAR, PR China
| | - Terence Kin-Wah Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, PR China
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, PR China.,State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong SAR, PR China
| | - Weiyuan John Kao
- Department of Industrial and Manufacturing Systems Engineering, Biomedical Engineering Program of Faculty of Engineering and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Regina Cheuk-Lam Lo
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, PR China.,State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong SAR, PR China
| |
Collapse
|
7
|
Gifford G, Stevenson W, Best G. Combination of the dual PIM/PI3-kinase inhibitor IBL-202 and venetoclax is effective in diffuse large B-cell lymphoma. Leuk Lymphoma 2020; 61:3165-3176. [PMID: 32723130 DOI: 10.1080/10428194.2020.1795156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Current chemoimmunotherapy is unable to cure up to 40% of patients diagnosed with diffuse large B-cell lymphoma (DLBCL). Targeting the mechanisms by which DLBCL evades apoptosis is crucial to overcoming treatment failure in this heterogeneous disease as both current and novel treatments depend on the apoptosis of malignant cells. Despite the common overexpression of BCL-2, venetoclax is ineffective in DLBCL due to MCL-1 co-expression. This is driven by pro-growth PI3-kinase signaling, which is promoted in turn by PIM kinases. In this study, the novel dual-kinase inhibitor, IBL-202, was combined with venetoclax against a panel of DLBCL cell lines that have variable expression of pro-survival proteins. The results support the efficacy of simultaneously targeting inter-related molecules to overcome apoptotic escape in this biologically heterogeneous disease. As venetoclax, pan-PIM-kinase and pan-PI3-kinase inhibitors have, or are currently being studied in clinical trials, it may be rational to consider these drugs in combination for the treatment of DLBCL.
Collapse
Affiliation(s)
- Grace Gifford
- Northern Blood Research Centre, Kolling Institution of Medical Research, The University of Sydney. St Leonards, Australia.,Department of Haematology, Royal North Shore Hospital, St Leonards, Australia
| | - William Stevenson
- Northern Blood Research Centre, Kolling Institution of Medical Research, The University of Sydney. St Leonards, Australia.,Department of Haematology, Royal North Shore Hospital, St Leonards, Australia
| | - Giles Best
- Northern Blood Research Centre, Kolling Institution of Medical Research, The University of Sydney. St Leonards, Australia.,Department of Haematology, Royal North Shore Hospital, St Leonards, Australia
| |
Collapse
|
8
|
Malone T, Schäfer L, Simon N, Heavey S, Cuffe S, Finn S, Moore G, Gately K. Current perspectives on targeting PIM kinases to overcome mechanisms of drug resistance and immune evasion in cancer. Pharmacol Ther 2019; 207:107454. [PMID: 31836451 DOI: 10.1016/j.pharmthera.2019.107454] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/03/2019] [Indexed: 12/22/2022]
Abstract
PIM kinases are a class of serine/threonine kinases that play a role in several of the hallmarks of cancer including cell cycle progression, metabolism, inflammation and immune evasion. Their constitutively active nature and unique catalytic structure has led them to be an attractive anticancer target through the use of small molecule inhibitors. This review highlights the enhanced activity of PIM kinases in cancer that can be driven by hypoxia in the tumour microenvironment and the important role that aberrant PIM kinase activity plays in resistance mechanisms to chemotherapy, radiotherapy, anti-angiogenic therapies and targeted therapies. We highlight an interaction of PIM kinases with numerous major oncogenic players, including but not limited to, stabilisation of p53, synergism with c-Myc, and notable parallel signalling with PI3K/Akt. We provide a comprehensive overview of PIM kinase's role as an escape mechanism to targeted therapies including PI3K/mTOR inhibitors, MET inhibitors, anti-HER2/EGFR treatments and the immunosuppressant rapamycin, providing a rationale for co-targeting treatment strategies for a more durable patient response. The current status of PIM kinase inhibitors and their use as a combination therapy with other targeted agents, in addition to the development of novel multi-molecularly targeted single therapeutic agents containing a PIM kinase targeting moiety are discussed.
Collapse
Affiliation(s)
- Tom Malone
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Lea Schäfer
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Nathalie Simon
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Sinead Cuffe
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Stephen Finn
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Gillian Moore
- School of Pharmacy and Biomolecular Sciences, RCSI, Dublin, Ireland
| | - Kathy Gately
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
9
|
Mohlin S, Hansson K, Radke K, Martinez S, Blanco-Apiricio C, Garcia-Ruiz C, Welinder C, Esfandyari J, O'Neill M, Pastor J, von Stedingk K, Bexell D. Anti-tumor effects of PIM/PI3K/mTOR triple kinase inhibitor IBL-302 in neuroblastoma. EMBO Mol Med 2019; 11:e10058. [PMID: 31310053 PMCID: PMC6685085 DOI: 10.15252/emmm.201810058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 06/04/2019] [Accepted: 06/24/2019] [Indexed: 11/12/2022] Open
Abstract
The PI3K pathway is a major driver of cancer progression. However, clinical resistance to PI3K inhibition is common. IBL‐302 is a novel highly specific triple PIM, PI3K, and mTOR inhibitor. Screening IBL‐302 in over 700 cell lines representing 47 tumor types identified neuroblastoma as a strong candidate for PIM/PI3K/mTOR inhibition. IBL‐302 was more effective than single PI3K inhibition in vitro, and IBL‐302 treatment of neuroblastoma patient‐derived xenograft (PDX) cells induced apoptosis, differentiated tumor cells, and decreased N‐Myc protein levels. IBL‐302 further enhanced the effect of the common cytotoxic chemotherapies cisplatin, doxorubicin, and etoposide. Global genome, proteome, and phospho‐proteome analyses identified crucial biological processes, including cell motility and apoptosis, targeted by IBL‐302 treatment. While IBL‐302 treatment alone reduced tumor growth in vivo, combination therapy with low‐dose cisplatin inhibited neuroblastoma PDX growth. Complementing conventional chemotherapy treatment with PIM/PI3K/mTOR inhibition has the potential to improve clinical outcomes and reduce severe late effects in children with high‐risk neuroblastoma.
Collapse
Affiliation(s)
- Sofie Mohlin
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Karin Hansson
- Department of Laboratory Medicine, Translational Cancer Research, Lund University Cancer Center, Lund University, Lund, Sweden
| | - Katarzyna Radke
- Department of Laboratory Medicine, Translational Cancer Research, Lund University Cancer Center, Lund University, Lund, Sweden
| | - Sonia Martinez
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Carmen Blanco-Apiricio
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cristian Garcia-Ruiz
- Department of Laboratory Medicine, Translational Cancer Research, Lund University Cancer Center, Lund University, Lund, Sweden
| | - Charlotte Welinder
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Javanshir Esfandyari
- Department of Laboratory Medicine, Translational Cancer Research, Lund University Cancer Center, Lund University, Lund, Sweden
| | | | - Joaquin Pastor
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Kristoffer von Stedingk
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Oncogenomics, University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Daniel Bexell
- Department of Laboratory Medicine, Translational Cancer Research, Lund University Cancer Center, Lund University, Lund, Sweden
| |
Collapse
|