1
|
Lin CHT, Tariq MJ, Ullah F, Sannareddy A, Khalid F, Abbas H, Bader A, Samaras C, Valent J, Khouri J, Anwer F, Raza S, Dima D. Current Novel Targeted Therapeutic Strategies in Multiple Myeloma. Int J Mol Sci 2024; 25:6192. [PMID: 38892379 PMCID: PMC11172591 DOI: 10.3390/ijms25116192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy caused by the clonal expansion of immunoglobulin-producing plasma cells in the bone marrow and/or extramedullary sites. Common manifestations of MM include anemia, renal dysfunction, infection, bone pain, hypercalcemia, and fatigue. Despite numerous recent advancements in the MM treatment paradigm, current therapies demonstrate limited long-term effectiveness and eventual disease relapse remains exceedingly common. Myeloma cells often develop drug resistance through clonal evolution and alterations of cellular signaling pathways. Therefore, continued research of new targets in MM is crucial to circumvent cumulative drug resistance, overcome treatment-limiting toxicities, and improve outcomes in this incurable disease. This article provides a comprehensive overview of the landscape of novel treatments and emerging therapies for MM grouped by molecular target. Molecular targets outlined include BCMA, GPRC5D, FcRH5, CD38, SLAMF7, BCL-2, kinesin spindle protein, protein disulfide isomerase 1, peptidylprolyl isomerase A, Sec61 translocon, and cyclin-dependent kinase 6. Immunomodulatory drugs, NK cell therapy, and proteolysis-targeting chimera are described as well.
Collapse
Affiliation(s)
- Cindy Hsin-Ti Lin
- Department of Internal Medicine, Case Western Reserve University, MetroHealth Campus, Cleveland, OH 44109, USA
| | - Muhammad Junaid Tariq
- Department of Hematology-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Fauzia Ullah
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | | | - Farhan Khalid
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA;
| | - Hasan Abbas
- Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Abbas Bader
- School of Medicine, University of Missouri–Kansas City, Kansas City, MO 64110, USA;
| | - Christy Samaras
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Jason Valent
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Jack Khouri
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Faiz Anwer
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Shahzad Raza
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Danai Dima
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
2
|
Kawaji-Kanayama Y, Kobayashi T, Muramatsu A, Uchiyama H, Sasaki N, Uoshima N, Nakao M, Takahashi R, Shimura K, Kaneko H, Kiyota M, Wada K, Chinen Y, Hirakawa K, Fuchida SI, Shimazaki C, Matsumura-Kimoto Y, Mizutani S, Tsukamoto T, Shimura Y, Horiike S, Taniwaki M, Kuroda J. Prognostic impact of resistance to bortezomib and/or lenalidomide in carfilzomib-based therapies for relapsed/refractory multiple myeloma: The Kyoto Clinical Hematology Study Group, multicenter, pilot, prospective, observational study in Asian patients. Cancer Rep (Hoboken) 2021; 5:e1476. [PMID: 34124862 PMCID: PMC8842705 DOI: 10.1002/cnr2.1476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/11/2022] Open
Abstract
Background Combinatory strategies with carfilzomib (CFZ), a second‐generation proteasome inhibitor, plus dexamethasone (DEX) with or without lenalidomide (LEN) have shown promising efficacy for patients with relapsed/refractory multiple myeloma (RRMM) in pivotal clinical trials. However, their effects on patients who were resistance to bortezomib (BTZ) and/or LEN have not been fully evaluated in a daily practice setting. Aims To evaluate the real‐world efficacy and safety of CFZ‐based treatments; that is, CFZ with LEN plus DEX (KRD therapy) and CFZ with DEX (KD therapy), in Asian patients, we conducted a multicenter pilot prospective observational study in the Kyoto Clinical Hematology Study Group. Methods and Results All 50 patients with RRMM enrolled in this study were treated with CFZ‐based treatments between 2017 and 2019. KRD and KD were administered to 31 and 19 patients, respectively. The overall response rates (ORRs) were 80.6% with KRD and 73.7% with KD. Two‐year progression‐free survival (PFS) and overall survival (OS) were 58.5% and 79.7% with KRD, and 23.1% and 52.6% with KD. By multivariate analysis, refractoriness to BTZ and to LEN were identified as independent unfavorable factors for both PFS and OS. The common non‐hematologic AEs included hypertension (42.0%), fever (24.0%), fatigue (24.0%), and infection (16.0%). No serious heart failure was observed. This study is registered as UMIN000025108. Conclusion This study suggests the need of the development of novel CFZ‐containing strategy which can overcome the refractoriness to BTZ and/or LEN, while both KRD and KD were shown to be mostly feasible in Asian patients in a daily practice setting.
Collapse
Affiliation(s)
- Yuka Kawaji-Kanayama
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsutomu Kobayashi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ayako Muramatsu
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Hematology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Hitoji Uchiyama
- Department of Hematology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Nana Sasaki
- Department of Hematology, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Nobuhiko Uoshima
- Department of Hematology, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Mitsushige Nakao
- Department of Internal Medicine, Otsu Municipal Hospital, Otsu, Japan
| | - Ryoichi Takahashi
- Department of Hematology, Omihachiman Community Medical Center, Omihachiman, Japan
| | - Kazuho Shimura
- Department of Hematology, Aiseikai Yamashina Hospital, Kyoto, Japan
| | - Hiroto Kaneko
- Department of Hematology, Aiseikai Yamashina Hospital, Kyoto, Japan
| | - Miki Kiyota
- Department of Hematology, Matsushita Memorial Hospital, Moriguchi, Japan
| | - Katsuya Wada
- Department of Hematology, Matsushita Memorial Hospital, Moriguchi, Japan
| | - Yoshiaki Chinen
- Department of Hematology, Fukuchiyama City Hospital, Fukuchiyama, Japan
| | - Koichi Hirakawa
- Department of Hematology, Fukuchiyama City Hospital, Fukuchiyama, Japan
| | - Shin-Ichi Fuchida
- Department of Hematology, Japan Community Health care Organization, Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Chihiro Shimazaki
- Department of Hematology, Japan Community Health care Organization, Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Yayoi Matsumura-Kimoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinsuke Mizutani
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taku Tsukamoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeo Horiike
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masafumi Taniwaki
- Department of Hematology, Aiseikai Yamashina Hospital, Kyoto, Japan.,Center for Molecular Diagnostic and Therapeutics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | |
Collapse
|
3
|
Algarín EM, Quwaider D, Campos-Laborie FJ, Díaz-Tejedor A, Mogollón P, Vuelta E, Martín-Sánchez M, San-Segundo L, González-Méndez L, Gutiérrez NC, García-Sanz R, Paíno T, De Las Rivas J, Ocio EM, Garayoa M. Stroma-Mediated Resistance to S63845 and Venetoclax through MCL-1 and BCL-2 Expression Changes Induced by miR-193b-3p and miR-21-5p Dysregulation in Multiple Myeloma. Cells 2021; 10:cells10030559. [PMID: 33806619 PMCID: PMC8001939 DOI: 10.3390/cells10030559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/20/2021] [Accepted: 02/27/2021] [Indexed: 12/18/2022] Open
Abstract
BH3-mimetics targeting anti-apoptotic proteins such as MCL-1 (S63845) or BCL-2 (venetoclax) are currently being evaluated as effective therapies for the treatment of multiple myeloma (MM). Interleukin 6, produced by mesenchymal stromal cells (MSCs), has been shown to modify the expression of anti-apoptotic proteins and their interaction with the pro-apoptotic BIM protein in MM cells. In this study, we assess the efficacy of S63845 and venetoclax in MM cells in direct co-culture with MSCs derived from MM patients (pMSCs) to identify additional mechanisms involved in the stroma-induced resistance to these agents. MicroRNAs miR-193b-3p and miR-21-5p emerged among the top deregulated miRNAs in myeloma cells when directly co-cultured with pMSCs, and we show their contribution to changes in MCL-1 and BCL-2 protein expression and in the activity of S63845 and venetoclax. Additionally, direct contact with pMSCs under S63845 and/or venetoclax treatment modifies myeloma cell dependence on different BCL-2 family anti-apoptotic proteins in relation to BIM, making myeloma cells more dependent on the non-targeted anti-apoptotic protein or BCL-XL. Finally, we show a potent effect of the combination of S63845 and venetoclax even in the presence of pMSCs, which supports this combinatorial approach for the treatment of MM.
Collapse
Affiliation(s)
- Esperanza M. Algarín
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Dalia Quwaider
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Francisco J. Campos-Laborie
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (F.J.C.-L.); (J.D.L.R.)
- The Gurdon Institute (Wellcome Trust/Cancer Research UK), University of Cambridge, Cambridge CB2 1QN, UK
| | - Andrea Díaz-Tejedor
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Elena Vuelta
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Montserrat Martín-Sánchez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Laura San-Segundo
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Lorena González-Méndez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Norma C. Gutiérrez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Ramón García-Sanz
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Teresa Paíno
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (F.J.C.-L.); (J.D.L.R.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39011 Santander, Spain;
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Correspondence: ; Tel.: +34-923-295812
| |
Collapse
|
4
|
Fuchs O. Treatment of Lymphoid and Myeloid Malignancies by Immunomodulatory Drugs. Cardiovasc Hematol Disord Drug Targets 2019; 19:51-78. [PMID: 29788898 DOI: 10.2174/1871529x18666180522073855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/05/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Thalidomide and its derivatives (lenalidomide, pomalidomide, avadomide, iberdomide hydrochoride, CC-885 and CC-90009) form the family of immunomodulatory drugs (IMiDs). Lenalidomide (CC5013, Revlimid®) was approved by the US FDA and the EMA for the treatment of multiple myeloma (MM) patients, low or intermediate-1 risk transfusion-dependent myelodysplastic syndrome (MDS) with chromosome 5q deletion [del(5q)] and relapsed and/or refractory mantle cell lymphoma following bortezomib. Lenalidomide has also been studied in clinical trials and has shown promising activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). Lenalidomide has anti-inflammatory effects and inhibits angiogenesis. Pomalidomide (CC4047, Imnovid® [EU], Pomalyst® [USA]) was approved for advanced MM insensitive to bortezomib and lenalidomide. Other IMiDs are in phases 1 and 2 of clinical trials. Cereblon (CRBN) seems to have an important role in IMiDs action in both lymphoid and myeloid hematological malignancies. Cereblon acts as the substrate receptor of a cullin-4 really interesting new gene (RING) E3 ubiquitin ligase CRL4CRBN. This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1α (CK1α) and marks them for degradation in proteasomes. Both these transcription factors (IKZF1 and IKZF3) stimulate proliferation of MM cells and inhibit T cells. Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Argonaute-2 seems to be an important drug target against IMiDs resistance in MM cells. Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. The CK1α gene (CSNK1A1) is located on 5q32 in commonly deleted region (CDR) in del(5q) MDS. Inhibition of CK1α sensitizes del(5q) MDS cells to lenalidomide. CK1α mediates also survival of malignant plasma cells in MM. Though, inhibition of CK1α is a potential novel therapy not only in del(5q) MDS but also in MM. High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. While transfusion independence (TI) after lenalidomide treatment is more than 60% in MDS patients with del(5q), only 25% TI and substantially shorter duration of response with occurrence of neutropenia and thrombocytopenia were achieved in lower risk MDS patients with normal karyotype treated with lenalidomide. Shortage of the biomarkers for lenalidomide response in these MDS patients is the main problem up to now.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| |
Collapse
|
5
|
Bosseler M, Marani V, Broukou A, Lequeux A, Kaoma T, Schlesser V, François JH, Palissot V, Berchem GJ, Aouali N, Janji B. Inhibition of HIF1α-Dependent Upregulation of Phospho-l-Plastin Resensitizes Multiple Myeloma Cells to Frontline Therapy. Int J Mol Sci 2018; 19:ijms19061551. [PMID: 29882856 PMCID: PMC6032243 DOI: 10.3390/ijms19061551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/08/2018] [Accepted: 05/12/2018] [Indexed: 12/18/2022] Open
Abstract
The introduction of novel frontline agents in multiple myeloma (MM), like immunomodulatory drugs and proteasome inhibitors, has improved the overall survival of patients. Yet, MM is still not curable, and drug resistance (DR) remains the main challenge. To improve the understanding of DR in MM, we established a resistant cell line (MOLP8/R). The exploration of DR mechanisms yielded an overexpression of HIF1α, due to impaired proteasome activity of MOLP8/R. We show that MOLP8/R, like other tumor cells, overexpressing HIF1α, have an increased resistance to the immune system. By exploring the main target genes regulated by HIF1α, we could not show an overexpression of these targets in MOLP8/R. We, however, show that MOLP8/R cells display a very high overexpression of LCP1 gene (l-Plastin) controlled by HIF1α, and that this overexpression also exists in MM patient samples. The l-Plastin activity is controlled by its phosphorylation in Ser5. We further show that the inhibition of l-Plastin phosphorylation restores the sensitivity of MOLP8/R to immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs). Our results reveal a new target gene of DR, controlled by HIF1α.
Collapse
Affiliation(s)
- Manon Bosseler
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Vanessa Marani
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Angelina Broukou
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Amandine Lequeux
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Tony Kaoma
- Bioinformatics and Modelling, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Vincent Schlesser
- Laboratory of Hematology, Centre Hospitalier de Luxembourg (CHL), L-1526 Luxembourg City, Luxembourg.
| | - Jean-Hugues François
- Laboratory of Hematology, Centre Hospitalier de Luxembourg (CHL), L-1526 Luxembourg City, Luxembourg.
| | - Valérie Palissot
- Laboratory of Oncolytic-Virus-Immuno-Therapeutics, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Guy J Berchem
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
- Laboratory of Hematology, Centre Hospitalier de Luxembourg (CHL), L-1526 Luxembourg City, Luxembourg.
| | - Nasséra Aouali
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Bassam Janji
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| |
Collapse
|
6
|
Hernández-García S, San-Segundo L, González-Méndez L, Corchete LA, Misiewicz-Krzeminska I, Martín-Sánchez M, López-Iglesias AA, Algarín EM, Mogollón P, Díaz-Tejedor A, Paíno T, Tunquist B, Mateos MV, Gutiérrez NC, Díaz-Rodriguez E, Garayoa M, Ocio EM. The kinesin spindle protein inhibitor filanesib enhances the activity of pomalidomide and dexamethasone in multiple myeloma. Haematologica 2017; 102:2113-2124. [PMID: 28860344 PMCID: PMC5709111 DOI: 10.3324/haematol.2017.168666] [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] [Received: 03/13/2017] [Accepted: 08/29/2017] [Indexed: 01/20/2023] Open
Abstract
Kinesin spindle protein inhibition is known to be an effective therapeutic approach in several malignancies. Filanesib (ARRY-520), an inhibitor of this protein, has demonstrated activity in heavily pre-treated multiple myeloma patients. The aim of the work herein was to investigate the activity of filanesib in combination with pomalidomide plus dexamethasone backbone, and the mechanisms underlying the potential synergistic effect. The ability of filanesib to enhance the activity of pomalidomide plus dexamethasone was studied in several in vitro and in vivo models. Mechanisms of this synergistic combination were dissected by gene expression profiling, immunostaining, cell cycle and short interfering ribonucleic acid studies. Filanesib showed in vitro, ex vivo, and in vivo synergy with pomalidomide plus dexamethasone treatment. Importantly, the in vivo synergy observed in this combination was more evident in large, highly proliferative tumors, and was shown to be mediated by the impairment of mitosis transcriptional control, an increase in monopolar spindles, cell cycle arrest and the induction of apoptosis in cells in proliferative phases. In addition, the triple combination increased the activation of the proapoptotic protein BAX, which has previously been associated with sensitivity to filanesib, and could potentially be used as a predictive biomarker of response to this combination. Our results provide preclinical evidence for the potential benefit of the combination of filanesib with pomalidomide and dexamethasone, and supported the initiation of a recently activated trial being conducted by the Spanish Myeloma group which is investigating this combination in relapsed myeloma patients.
Collapse
Affiliation(s)
- Susana Hernández-García
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Laura San-Segundo
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Lorena González-Méndez
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Luis A Corchete
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Irena Misiewicz-Krzeminska
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain.,National Medicines Institute, Warsaw, Poland
| | - Montserrat Martín-Sánchez
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Ana-Alicia López-Iglesias
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | | | - Pedro Mogollón
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Andrea Díaz-Tejedor
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Teresa Paíno
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | | | - María-Victoria Mateos
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Norma C Gutiérrez
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Elena Díaz-Rodriguez
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Mercedes Garayoa
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Enrique M Ocio
- Centro Investigación del Cáncer-IBMCC (CSIC-USAL) and Hospital Universitario-IBSAL, Salamanca, Spain
| |
Collapse
|
7
|
Clemens J, Welti L, Schäfer J, Seckinger A, Burhenne J, Theile D, Weiss J. Bortezomib, carfilzomib and ixazomib do not mediate relevant transporter-based drug-drug interactions. Oncol Lett 2017; 14:3185-3192. [PMID: 28927064 DOI: 10.3892/ol.2017.6560] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/09/2017] [Indexed: 01/14/2023] Open
Abstract
In order to optimize the clinical application of an increasing number of proteasome inhibitors, investigations into the differences between their respective pharmacodynamic and pharmacokinetic profiles, including their ability to act as a perpetrator in drug-drug interactions, are warranted. Therefore, in the present in vitro study, it was investigated whether bortezomib, carfilzomib and ixazomib are able to alter the expression, and/or the activity, of specific drug transporters generally relevant for pharmacokinetic drug-drug interactions. Through induction experiments, the current study demonstrated that the aforementioned three proteasome inhibitors do not induce mRNA expression of the transporter genes ATP binding cassette (ABC)B1, C1, C2 and G2 in the LS180 cell line, which was used as a model for systemic induction. By contrast, in certain myeloma cell lines, ixazomib provoked minor alterations in individual transporter gene expression. None of the proteasome inhibitors tested relevantly inhibited drug transporters within the range of physiological plasma concentrations. Taken together, transporter-based drug-drug interactions are unlikely to be a primary concern in the clinical application of the tested compounds.
Collapse
Affiliation(s)
- Jannick Clemens
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Lukas Welti
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Julia Schäfer
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Anja Seckinger
- Department of Internal Medicine V, Oncology, Hematology and Rheumatology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Dirk Theile
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
8
|
Schönfeld K, Zuber C, Pinkas J, Häder T, Bernöster K, Uherek C. Indatuximab ravtansine (BT062) combination treatment in multiple myeloma: pre-clinical studies. J Hematol Oncol 2017; 10:13. [PMID: 28077160 PMCID: PMC5225632 DOI: 10.1186/s13045-016-0380-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 12/26/2016] [Indexed: 01/16/2023] Open
Abstract
Indatuximab ravtansine is a monoclonal antibody-linked cytotoxic agent that specifically targets CD138-expressing cells. Monotherapy has been shown to significantly inhibit multiple myeloma tumour growth in vivo and improve host survival. Here, we show that in most cell lines tested, indatuximab ravtansine acts additively or even synergistically with clinically approved therapies for treatment of multiple myeloma. In addition, in vivo mouse xenograft models confirmed the activity of indatuximab ravtansine in combination with lenalidamide and lenalidomide/dexamethasone. Indatuximab ravtansine may therefore be a suitable combination partner for multiple myeloma, and a clinical study is ongoing.
Collapse
Affiliation(s)
- Kurt Schönfeld
- Biotest AG, Landsteinerstraße 5, 63303, Dreieich, Germany
| | - Chantal Zuber
- Biotest AG, Landsteinerstraße 5, 63303, Dreieich, Germany
| | - Jan Pinkas
- ImmunoGen Inc., 830 Winter Street, Waltham, 02451-1477, MA, USA
| | - Thomas Häder
- Biotest AG, Landsteinerstraße 5, 63303, Dreieich, Germany
| | | | | |
Collapse
|
9
|
Burger R, Günther A, Klausz K, Staudinger M, Peipp M, Penas EMM, Rose-John S, Wijdenes J, Gramatzki M. Due to interleukin-6 type cytokine redundancy only glycoprotein 130 receptor blockade efficiently inhibits myeloma growth. Haematologica 2016; 102:381-390. [PMID: 27658435 DOI: 10.3324/haematol.2016.145060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/14/2016] [Indexed: 12/25/2022] Open
Abstract
Interleukin-6 has an important role in the pathophysiology of multiple myeloma where it supports the growth and survival of the malignant plasma cells in the bone marrow. It belongs to a family of cytokines which use the glycoprotein 130 chain for signal transduction, such as oncostatin M or leukemia inhibitory factor. Targeting interleukin-6 in plasma cell diseases is currently evaluated in clinical trials with monoclonal antibodies. Here, efforts were made to elucidate the contribution of interleukin-6 and glycoprotein 130 signaling in malignant plasma cell growth in vivo In the xenograft severe combined immune deficiency model employing our interleukin-6-dependent plasma cell line INA-6, the lack of human interleukin-6 induced autocrine interleukin-6 production and a proliferative response to other cytokines of the glycoprotein 130 family. Herein, mice were treated with monoclonal antibodies against human interleukin-6 (elsilimomab/B-E8), the interleukin-6 receptor (B-R6), and with an antibody blocking glycoprotein 130 (B-R3). While treatment of mice with interleukin-6 and interleukin-6 receptor antibodies resulted in a modest delay in tumor growth, the development of plasmacytomas was completely prevented with the anti-glycoprotein 130 antibody. Importantly, complete inhibition was also achieved using F(ab')2-fragments of monoclonal antibody B-R3. Tumors harbor activated signal transducer and activator of transcription 3, and in vitro, the antibody inhibited leukemia inhibitory factor stimulated signal transducer and activator of transcription 3 phosphorylation and cell growth, while being less effective against interleukin-6. In conclusion, the growth of INA-6 plasmacytomas in vivo under interleukin-6 withdrawal remains strictly dependent on glycoprotein 130, and other glycoprotein 130 cytokines may substitute for interleukin-6. Antibodies against glycoprotein 130 are able to overcome this redundancy and should be explored for a possible therapeutic window.
Collapse
Affiliation(s)
- Renate Burger
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Andreas Günther
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Katja Klausz
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Matthias Staudinger
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Eva Maria Murga Penas
- Institute of Human Genetics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Stefan Rose-John
- Department of Biochemistry, Christian-Albrechts-University of Kiel, Medical Faculty, Germany
| | | | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
10
|
Manasanch EE, de Larrea CF, Zingone A, Steinberg SM, Kwok M, Tageja N, Bhutani M, Kazandjian D, Roschewski M, Wu P, Carter G, Zuchlinski D, Mulquin M, Lamping L, Costello R, Burton D, Gil LA, Figg WD, Maric I, Calvo KR, Yuan C, Stetler-Stevenson M, Korde N, Landgren O. Enzymatic activities of circulating plasma proteasomes in newly diagnosed multiple myeloma patients treated with carfilzomib, lenalidomide and dexamethasone. Leuk Lymphoma 2016; 58:639-645. [PMID: 27687480 DOI: 10.1080/10428194.2016.1214953] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The proteasome inhibitor carfilzomib is highly effective in the treatment of multiple myeloma. It irreversibly binds the chymotrypsin-like active site in the β5 subunit of the 20S proteasome. Despite impressive response rates when carfilzomib is used in combination with immunomodulatory agents in newly diagnosed multiple myeloma patients; no biomarker exists to accurately predict response and clinical outcomes. We prospectively assessed the activity in peripheral blood of the chymotrypsin-like (CHYM), caspase-like (CASP) and trypsin-like (TRYP) proteolytic sites in 45 newly diagnosed multiple myeloma patients treated with eight cycles of carfilzomib, lenalidomide and dexamethasone (CRd) (NCT01402284). Samples were collected per protocol and proteasome activity measured through a fluorogenic assay. Median CHYM levels after one dose of carfilzomib decreased by >70%. CHYM and CASP activity decreased throughout treatment reaching a minimum after eight cycles of treatment. Higher levels of proteasome activity associated with higher disease burden (r > 0.30; p < 0.05) and higher disease stage (0.10 < p <0.20). No association was found with the probability of achieving a complete response, minimal residual disease negativity or time to best response. Further studies evaluating proteasome activity in malignant plasma cells may help elucidate how proteasome activity can be used as a biomarker in multiple myeloma.
Collapse
Affiliation(s)
- Elisabet E Manasanch
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA.,b Department of Lymphoma/Myeloma, Division of Cancer Medicine , University of Texas, M.D. Anderson Cancer Center , Houston , TX , USA
| | - Carlos Fernández de Larrea
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA.,c Amyloidosis and Myeloma Unit, Department of Hematology , Hospital Clínic, Barcelona. Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona , Barcelona , Catalonia , Spain
| | - Adriana Zingone
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Seth M Steinberg
- d Biostastistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Mary Kwok
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Nishant Tageja
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Manisha Bhutani
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Dickran Kazandjian
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Mark Roschewski
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Peter Wu
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - George Carter
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Diamond Zuchlinski
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Marcia Mulquin
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Liz Lamping
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Rene Costello
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Deborah Burton
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Lindsay A Gil
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - William D Figg
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Irina Maric
- e Laboratory of Hematology, Department of Laboratory Medicine , NIH Clinical Center , Bethesda , MD , USA
| | - Katherine R Calvo
- e Laboratory of Hematology, Department of Laboratory Medicine , NIH Clinical Center , Bethesda , MD , USA
| | - Constance Yuan
- f Flow Cytometry Unit, Laboratory of Pathology, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Maryalice Stetler-Stevenson
- f Flow Cytometry Unit, Laboratory of Pathology, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Neha Korde
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA.,g Myeloma Service, Department of Medicine , Memorial Sloan-Kettering Cancer Center , New York , NY , USA
| | - Ola Landgren
- a Multiple Myeloma Section , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA.,g Myeloma Service, Department of Medicine , Memorial Sloan-Kettering Cancer Center , New York , NY , USA
| |
Collapse
|
11
|
Integrated analysis of microRNAs, transcription factors and target genes expression discloses a specific molecular architecture of hyperdiploid multiple myeloma. Oncotarget 2016; 6:19132-47. [PMID: 26056083 PMCID: PMC4662480 DOI: 10.18632/oncotarget.4302] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/13/2015] [Indexed: 12/13/2022] Open
Abstract
Multiple Myeloma (MM) is a malignancy characterized by the hyperdiploid (HD-MM) and the non-hyperdiploid (nHD-MM) subtypes. To shed light within the molecular architecture of these subtypes, we used a novel integromics approach. By annotated MM patient mRNA/microRNA (miRNA) datasets, we investigated mRNAs and miRNAs profiles with relation to changes in transcriptional regulators expression. We found that HD-MM displays specific gene and miRNA expression profiles, involving the Signal Transducer and Activator of Transcription (STAT)3 pathway as well as the Transforming Growth Factor–beta (TGFβ) and the transcription regulator Nuclear Protein-1 (NUPR1). Our data define specific molecular features of HD-MM that may translate in the identification of novel relevant druggable targets.
Collapse
|
12
|
[Advances on immunomodulatory drugs against multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 37:262-4. [PMID: 27033772 PMCID: PMC7342946 DOI: 10.3760/cma.j.issn.0253-2727.2016.03.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Dai Y, Jin F. Novel mechanisms of action for immunomodulatory drugs (IMiDs) against multiple myeloma: from a tragedy to a therapy. ACTA ACUST UNITED AC 2016. [DOI: 10.15436/2381-1404.15.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
14
|
Ogo N, Ishikawa Y, Sawada JI, Matsuno K, Hashimoto A, Asai A. Structure-Guided Design of Novel l-Cysteine Derivatives as Potent KSP Inhibitors. ACS Med Chem Lett 2015; 6:1004-9. [PMID: 26396688 DOI: 10.1021/acsmedchemlett.5b00221] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/22/2015] [Indexed: 12/31/2022] Open
Abstract
Kinesin spindle protein (KSP), known as Hs Eg5, a member of the kinesin-5 family, plays an important role in the formation and maintenance of the bipolar spindle. We previously reported S-trityl-l-cysteine derivatives as selective KSP inhibitors. Here, we report further optimizations using docking modeling in the L5 allosteric binding site, which led to the discovery of several high affinity derivatives with two fused phenyl rings in the trityl group giving low nanomolar range KSP ATPase inhibition. The representative derivatives potently inhibited cell growth of HCT116 cells in correlation with KSP inhibitory activities and significantly suppressed tumor growth in the xenograft model in vivo.
Collapse
Affiliation(s)
- Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical
Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Yoshinobu Ishikawa
- Department of Physical Biochemistry, School
of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Jun-ichi Sawada
- Center for Drug Discovery, Graduate School of Pharmaceutical
Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Kenji Matsuno
- Center for Drug Discovery, Graduate School of Pharmaceutical
Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Akihiro Hashimoto
- Tsukuba Research Center, Taiho Pharmaceutical Co., Ltd., 3 Okubo, Tsukuba, Ibaraki 300-2611, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical
Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| |
Collapse
|
15
|
Guidelines for determination of the number of prior lines of therapy in multiple myeloma. Blood 2015; 126:921-2. [DOI: 10.1182/blood-2015-05-647636] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
16
|
|
17
|
Jordan SC, Choi J, Vo A. Achieving incompatible transplantation through desensitization: current perspectives and future directions. Immunotherapy 2015; 7:377-98. [DOI: 10.2217/imt.15.10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The application of life-saving transplantation is severely limited by the shortage of organs, and histoincompatibility. To increase transplant rates in sensitized patients, new protocols for HLA and blood type incompatible (ABOi) desensitization have emerged. These approaches require significant desensitization using intravenous immunoglobulin, rituximab and plasma exchange. In addition, the development of donor-specific antibody responses post transplant is the major cause of allograft failure with return to dialysis. This increases patient morbidity/mortality and cost. Immunotherapeutic agents used for desensitization evolved from drug development in oncology and autoimmune diseases. Currently, there is a renaissance in development of novel drugs likely to improve antibody reduction in transplantation. These include agents that inactivate IgG molecules, anticytokine antibodies, costimulatory molecule blockade, anticomplement agents and therapies aimed at the plasma cell.
Collapse
Affiliation(s)
- Stanley C Jordan
- Comprehensive Transplant Center, Kidney Transplant Program & Transplant Immunotherapy Program, Cedars-Sinai Medical Center, 8900 Beverly Blvd, Los Angeles, CA 90048, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Nephrology & Transplant Immunology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jua Choi
- Comprehensive Transplant Center, Kidney Transplant Program & Transplant Immunotherapy Program, Cedars-Sinai Medical Center, 8900 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Ashley Vo
- Comprehensive Transplant Center, Kidney Transplant Program & Transplant Immunotherapy Program, Cedars-Sinai Medical Center, 8900 Beverly Blvd, Los Angeles, CA 90048, USA
| |
Collapse
|
18
|
Richardson PG, Moreau P, Laubach JP, Gupta N, Hui AM, Anderson KC, San Miguel JF, Kumar S. The investigational proteasome inhibitor ixazomib for the treatment of multiple myeloma. Future Oncol 2015; 11:1153-68. [DOI: 10.2217/fon.15.9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
ABSTRACT Ixazomib is an investigational, reversible 20S proteasome inhibitor. It is the first oral proteasome inhibitor under clinical investigation in multiple myeloma (MM). Under physiological conditions, the stable citrate ester drug substance, ixazomib citrate (MLN9708), rapidly hydrolyzes to the biologically active boronic acid, ixazomib (MLN2238). Preclinical studies have demonstrated antitumor activity in MM cell lines and xenograft models. In Phase I/II clinical studies ixazomib has had generally manageable toxicities, with limited peripheral neuropathy observed to date. Preliminary data from these studies indicate ixazomib is active as a single agent in relapsed/refractory MM and as part of combination regimens in newly diagnosed patients. Phase III studies in combination with lenalidomide–dexamethasone are ongoing.
Collapse
Affiliation(s)
- Paul G Richardson
- Division of Hematologic Malignancy, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Philippe Moreau
- Hematology Department, University Hospital Hotel-Dieu, Nantes, France
| | - Jacob P Laubach
- Division of Hematologic Malignancy, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Neeraj Gupta
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Ltd, Cambridge, MA, USA
| | - Ai-Min Hui
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Ltd, Cambridge, MA, USA
| | - Kenneth C Anderson
- Division of Hematologic Malignancy, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Jesús F San Miguel
- Clinica Universidad de Navarra, Centro Investigación Medica Aplicada (CIMA), Pamplona, Spain
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
19
|
Hansson M, Gimsing P, Badros A, Niskanen TM, Nahi H, Offner F, Salomo M, Sonesson E, Mau-Sorensen M, Stenberg Y, Sundberg A, Teige I, Van Droogenbroeck J, Wichert S, Zangari M, Frendeus B, Korsgren M, Poelman M, Tricot G. A Phase I Dose-Escalation Study of Antibody BI-505 in Relapsed/Refractory Multiple Myeloma. Clin Cancer Res 2015; 21:2730-6. [DOI: 10.1158/1078-0432.ccr-14-3090] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/03/2015] [Indexed: 11/16/2022]
|
20
|
Abstract
Multiple myeloma (MM) is a plasma cell malignancy leading to significant life-expectancy shortening. Although the incorporation of the novel agents thalidomide, bortezomib, and lenalidomide in the front-line therapy has resulted in significant improvement, almost all patients relapse, making the treatment of relapse a real challenge. In the present article, when and how to treat relapsed MM is discussed. Treatment can be safely delayed in a subset of patients with asymptomatic relapse, whereas those with symptomatic relapse, advanced disease at diagnosis, or significant paraproteinemic increase require prompt rescue therapy. The benefit of retreatment and the use of a sequential approach for successive relapses considering drug synergism are highlighted. For patients with aggressive relapses and for those who have exhausted all available options, continued therapy until disease progression is recommended, particularly when using regimens with a long-term safety profile. Patients with a duration response to a first autologous stem cell transplantation (ASCT) longer than 2 years may benefit from a second ASCT. Patients with aggressive disease and/or poor cytogenetics at diagnosis relapsing within the first 2 years from ASCT should be considered for an allogeneic transplantation. Finally, a number of newer promising drugs are being actively investigated and the enrolment of patients in clinical trials is encouraged.
Collapse
|
21
|
Leotta M, Biamonte L, Raimondi L, Ronchetti D, Di Martino MT, Botta C, Leone E, Pitari MR, Neri A, Giordano A, Tagliaferri P, Tassone P, Amodio N. A p53-dependent tumor suppressor network is induced by selective miR-125a-5p inhibition in multiple myeloma cells. J Cell Physiol 2014; 229:2106-16. [PMID: 24819167 DOI: 10.1002/jcp.24669] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 03/31/2014] [Accepted: 05/09/2014] [Indexed: 02/06/2023]
Abstract
The analysis of deregulated microRNAs (miRNAs) is emerging as a novel approach to disclose the regulation of tumor suppressor or tumor promoting pathways in tumor cells. Targeting aberrantly expressed miRNAs is therefore a promising strategy for cancer treatment. By miRNA profiling of primary plasma cells from multiple myeloma (MM) patients, we previously reported increased miR-125a-5p levels associated to specific molecular subgroups. On these premises, we aimed at investigating the biological effects triggered by miR-125a-5p modulation in MM cells. Expression of p53 pathway-related genes was down-regulated in MM cells transfected with miR-125a-5p mimics. Luciferase reporter assays confirmed specific p53 targeting at 3'UTR level by miR-125a-5p mimics. Interestingly, bone marrow stromal cells (BMSCs) affected the miR-125a-5p/p53 axis, since adhesion of MM cells to BMSCs strongly up-regulated miR-125a-5p levels, while reduced p53 expression. Moreover, ectopic miR-125a-5p reduced, while miR-125-5p inhibitors promoted, the expression of tumor suppressor miR-192 and miR-194, transcriptionally regulated by p53. Lentiviral-mediated stable inhibition of miR-125a-5p expression in wild-type p53 MM cells dampened cell growth, increased apoptosis and reduced cell migration. Importantly, inhibition of in vitro MM cell proliferation and migration was also achieved by synthetic miR-125a-5p inhibitors and was potentiated by the co-expression of miR-192 or miR-194. Taken together, our data indicate that miR-125a-5p antagonism results in the activation of p53 pathway in MM cells, underlying the crucial role of this miRNA in the biopathology of MM and providing the molecular rationale for the combinatory use of miR-125a inhibitors and miR-192 or miR-194 mimics for MM treatment.
Collapse
Affiliation(s)
- Marzia Leotta
- Department of Experimental and Clinical Medicine, Magna Graecia University, Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Identifying Professional Education Gaps and Barriers in Multiple Myeloma Patient Care: Findings of the Managing Myeloma Continuing Educational Initiative Advisory Committee. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2014; 14:356-69. [DOI: 10.1016/j.clml.2014.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 03/26/2014] [Accepted: 04/03/2014] [Indexed: 12/31/2022]
|
23
|
Richardson PG, Bladé J. The comprehensive clinical management of multiple myeloma and related-plasma cell disorders. Expert Rev Hematol 2014; 7:1-3. [PMID: 24483345 DOI: 10.1586/17474086.2014.882763] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Whilst we think of multiple myeloma and related plasma cell disorders as incurable to date, never before has there been such hope and enthusiasm about advances in the research and treatment of these various diseases. Translational research is very much at the forefront of progress for further refining targeted therapies and continuing to improve clinical efficacy. Whilst some of these advances in the last decade have been truly dramatic in their scope and timing, it is also worth noting that relatively incremental changes have favorably impacted on patient outcome, and this comprehensive clinical management review captures these accordingly. We hope therefore that this concise overview will give readers, be they specialist hemato-oncologists, or other providers and researchers in the field, an enlightening insight into the exciting future of therapeutic opportunities, as well as a practical 'hands on' approach.
Collapse
Affiliation(s)
- Paul G Richardson
- RJ Corman Professor of Medicine, Harvard Medical School Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Boston, MA, USA
| | | |
Collapse
|
24
|
Resistance to Proteasome Inhibitors in Multiple Myeloma. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2014. [DOI: 10.1007/978-3-319-06752-0_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|