1
|
Carlisi M, Lo Presti R, Spoto C, Mancuso S, Siragusa S, Caimi G. Calculated haemorheological profile and laboratory parameters in new diagnosed multiple myeloma patients: retrospective analysis according to survival. Ther Adv Hematol 2024; 15:20406207241298865. [PMID: 39583654 PMCID: PMC11585042 DOI: 10.1177/20406207241298865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/14/2024] [Indexed: 11/26/2024] Open
Abstract
Background Multiple myeloma is a malignant haematological neoplasm characterised by clonal proliferation of plasma cells, with a complex clinical picture, and a significant impact on patient survival, in which the prognosis evaluation of patients is of great importance. Objectives In this single-centre retrospective analysis, performed in a group of newly diagnosed multiple myeloma patients, we examined several clinical and laboratory parameters in order to evaluate their trend according to survival of patients. Design We collected data from 190 newly diagnosed multiple myeloma evaluated at the Hematology Division of the 'Paolo Giaccone' University Hospital of Palermo from 1 January 2017 to 30 September 2022. Specifically, we performed our analysis in the entire cohort of patients and also in the specific disease isotype. Methods We evaluated simple and low-cost laboratory and haemorheological parameters, the latter obtained in a calculated way. The primary endpoint was to evaluate the trend and the differences of these parameters in the study population, divided into two specific groups, deceased and survivors after a specific observation period of almost 7 years. Results In the entire cohort of multiple myeloma patients, we observed a mortality rate of 40%, of whom 36.4% were men and 43.1% were women. Among the patients who died, in comparison with those who survived, it is significantly evident the increase in age, in red cell distribution width (RDW), RDW%/albumin ratio and in the RDW%/haemoglobin ratio; moreover, in the same patients subgroup, we observed a reduction in haematocrit, total serum protein, calculated whole blood viscosity (evaluated according to the de Simone formula), serum albumin, albumin/fibrinogen ratio and in haemoglobin levels. Conclusion The obtained data can represent a possible starting point for subsequent targeted analyses, aimed at studying the prognostic value of each individual parameter considered, favouring an increasingly complete and immediate prognostic evaluation of patients.
Collapse
Affiliation(s)
- Melania Carlisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Via del Vespro 129, Palermo 90127, Italy
| | - Rosalia Lo Presti
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Corinne Spoto
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Salvatrice Mancuso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Sergio Siragusa
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Gregorio Caimi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
2
|
Li M, Wu H, Shou C, Peng Y, Song X, Ying W, Chen Y, Tong X. Optimal cut-off values and diagnostic significance of clinical laboratory indicators in newly diagnosed multiple myeloma. Discov Oncol 2024; 15:477. [PMID: 39331239 PMCID: PMC11436520 DOI: 10.1007/s12672-024-01254-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/20/2024] [Indexed: 09/28/2024] Open
Abstract
OBJECTIVE This study aims to identify clinical laboratory parameters for the diagnosis of newly diagnosed multiple myeloma (NDMM), establish optimal cutoffs for early screening, and develop a diagnostic model for precise diagnosis. METHODS The study conducted a retrospective analysis of 279 NDMM patients and 553 healthy subjects at Zhejiang Province People's Hospital between January 2008 and June 2023. Multifactor LR was employed to explore clinical laboratory indicators with diagnostic value for NDMM, determine optimal cutoff values and contract a diagnostic model. The diagnostic efficacy and clinical utility were evaluated using receiver operating characteristic curves (ROC), sensitivity, specificity, and other indicators. RESULTS Multifactor analysis revealed that hemoglobin (Hb), albumin (Alb), and platelet distribution width (PDW) were significant diagnostic factors for NDMM. Optimal cutoff values for Hb, Alb, and PDW in MM diagnosis were determined, and the results showed a significant increase in the probability of NDMM diagnosis when Alb was below 39.3 g/L, Hb was below 11.6 g/dL, and PDW was below 14.1 fL. The diagnostic model constructed from the development cohort demonstrated a high area under the ROC curve of 0.960 (95% CI 0.942-0.978) and exhibited good sensitivity (0.860), specificity (0.957). The area under the curve (AUC) value of the diagnostic model in the external validation cohort was 0.979, confirming its good diagnostic efficacy and generalization. CONCLUSIONS The optimal cutoff values for Hb, Alb, and PDW and the diagnostic model designed in the study provided good accuracy and sensitivity for the initial screening and diagnosis of NDMM.
Collapse
Affiliation(s)
- Manning Li
- Graduate School of Clinical Medicine, Jinzhou Medical University, 40 Songpo Road, Jinzhou, 121001, Liaoning, China
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Han Wu
- Graduate School of Clinical Medicine, Jinzhou Medical University, 40 Songpo Road, Jinzhou, 121001, Liaoning, China
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Chunyi Shou
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Ye Peng
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Xiaolu Song
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Wang Ying
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Xihu University, 261 Huansha Road, Hangzhou, 310006, Zhejiang, People's Republic of China.
| | - Yirui Chen
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China.
| | - Xiangmin Tong
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China.
- Department of Hematology, Affiliated Hangzhou First People's Hospital, Xihu University, 261 Huansha Road, Hangzhou, 310006, Zhejiang, People's Republic of China.
| |
Collapse
|
3
|
Bruzzese A, Martino EA, Mendicino F, Lucia E, Olivito V, Capodanno I, Neri A, Morabito F, Vigna E, Gentile M. Myelodysplastic syndromes del(5q): Pathogenesis and its therapeutic implications. Eur J Haematol 2024; 112:860-869. [PMID: 38294126 DOI: 10.1111/ejh.14181] [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: 12/14/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
Myelodysplastic syndromes (MDS) encompass a heterogeneous set of acquired bone marrow neoplastic disorders characterized by ineffective hematopoiesis within one or more bone marrow lineages. Nearly half of MDS patients carry cytogenetic alterations, with del(5q) being the most prevalent. Since its first description, del(5q) was consistently correlated with a typical clinical phenotype marked by anemia, thrombocytosis, and a low risk of evolving into acute leukemia. Presently, the World Health Organization (WHO) classification of myeloid neoplasms recognizes a specific subtype of MDS known as "myelodysplastic neoplasm with low blast and isolated del(5q)" identified by the sole presence of 5q deletion or in combination with one other abnormality excluding -7/del(7q). Several studies have sought to unravel the biological processes triggered by del(5q) in the development of MDS, revealing the involvement of various genes localized in specific regions of chromosome 5 referred to as common deleted regions (CDR). This intricate biological landscape makes the MDS cells with del(5q) exceptionally sensitive to lenalidomide. Several studies have confirmed the efficacy of lenalidomide in this context. Regrettably, the response to lenalidomide is not conclusive, prompting ongoing research into biological mechanisms that drive patients toward leukemia and strategies to circumvent lenalidomide resistance and disease progression.
Collapse
Affiliation(s)
- Antonella Bruzzese
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | | | - Francesco Mendicino
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Eugenio Lucia
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Virginia Olivito
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | | | - Antonino Neri
- Scientific Direction Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Fortunato Morabito
- Biotechnology Research Unit, Aprigliano, A.O./ASP of Cosenza, Cosenza, Italy
| | - Ernesto Vigna
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Massimo Gentile
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
4
|
Lecot L, Desmas‐Bazelle I, Benjamin S, De Fornel P, Ponce F, Kornya M, Desquilbet L, Beaudu‐Lange C, Ibisch C, Sayag D, Benchekroun G, Béguin J. Descriptive analysis and prognostic factors in cats with myeloma-related disorders: A multicenter retrospective study of 50 cases. J Vet Intern Med 2024; 38:1693-1705. [PMID: 38517293 PMCID: PMC11099758 DOI: 10.1111/jvim.17051] [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: 09/05/2023] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Myeloma-related disorders (MRDs) are rare and poorly documented neoplasms of cats. HYPOTHESIS/OBJECTIVES To describe clinical, clinicopathologic, and imaging findings, response to treatment, and survival time and to identify factors associated with shorter outcomes in cats with MRD. ANIMALS Fifty cats with a diagnosis of MRD. METHODS Cats with paraproteinemia confirmed by serum protein electrophoresis (SPE) and either intramedullary plasmacytosis >10%, marked cytonuclear atypia with intramedullary plasmacytosis that ranged between 5% and 10%, or cytologically or histologically confirmed visceral infiltration were retrospectively included from several veterinary referral centers. RESULTS Bone marrow plasmacytosis and splenic or hepatic involvement were present in 17/27 cats (63%), 36/42 cats (86%), and 27/38 cats (71%), respectively. Anemia was reported in 33/49 cats (67%) and thrombocytopenia in 16/47 cats (34%). Some of the treatments that the cats received included melphalan and prednisolone (n = 19), cyclophosphamide and prednisolone (n = 10), chlorambucil and prednisolone (n = 4), prednisolone (n = 4), or other (n = 4). The overall response rates to melphalan, cyclophosphamide, and chlorambucil in combination with prednisolone were 87%, 90%, and 100%, respectively. Adverse events to melphalan or cyclophosphamide occurred in 65% and 23% of cats, respectively. Median survival time was 122 days (range, 0-1403) and was not significantly associated with chemotherapy protocol. Anemia (hazard ratio [HR], 3.1; 95% confidence interval [CI], 1.0-9.8) and thrombocytopenia (HR, 2.7; 95% CI, 1.2-6.0) were risk factors for shorter survival. CONCLUSIONS AND CLINICAL IMPORTANCE Our study confirmed the guarded prognosis of MRD in cats and identified risk factors for shorter survival times.
Collapse
Affiliation(s)
- Lorris Lecot
- Ecole Nationale Vétérinaire d'Alfort–CHUVAService de Médecine InterneMaisons‐AlfortFrance
- Université de Lyon, VetAgro Sup, Service de médecine interneMarcy l'EtoileFrance
| | | | | | | | - Frédérique Ponce
- Université de Lyon, VetAgro Sup, Service de cancérologie, UR ICEMarcy l'EtoileFrance
| | - Matthew Kornya
- Ontario Veterinary CollegeUniversity of GuelphGuelphOntarioCanada
| | - Loïc Desquilbet
- Ecole nationale vétérinaire d'Alfort, IMRBMaisons‐AlfortFrance
| | | | - Catherine Ibisch
- Nantes‐Atlantic College of Veterinary Medicine and Food Sciences (Oniris)NantesFrance
| | | | - Ghita Benchekroun
- Ecole Nationale Vétérinaire d'Alfort–CHUVAService de Médecine InterneMaisons‐AlfortFrance
- Ecole Nationale Vétérinaire d'AlfortUniversité Paris Est Créteil, INSERM, IMRBMaisons‐AlfortFrance
| | - Jérémy Béguin
- Ecole Nationale Vétérinaire d'Alfort–CHUVAService de Médecine InterneMaisons‐AlfortFrance
- UMR1161 VIROLOGIE, INRAE, Ecole Nationale Vétérinaire d'Alfort, ANSESUniversité Paris‐EstMaisons‐AlfortFrance
| |
Collapse
|
5
|
Hou L, Li Y, Kang L, Li X, Li H, Xue F. The long-term mediation role of cytokines on the causal pathway from maternal gestational age to offspring eye diseases: Lifecourse-Network Mendelian randomization. Int Immunopharmacol 2023; 122:110667. [PMID: 37487263 DOI: 10.1016/j.intimp.2023.110667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/28/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Gestational duration has a significant impact on eye diseases. A large number of evidences suggest that cytokines are associated with gestational duration and eye diseases. However, the causal relationships among cytokines, maternal gestational impairment and offspring eye diseases remain unclear. METHODS We performed lifecourse-network Mendelian randomization (MR) to explore the causal relationships between maternal gestational duration (from the Early Growth Genetics (EGG) Consortium and the Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH) study, N = 84,689), neonatal/adult cytokines (from the NHGRI-EBI Catalog, N = 764/4,618), and adult eye diseases (from FinnGen consotium, N = 309,154) using summary-level data from large genome-wide association studies. Multiplicative random effects inverse variance weighted (IVW) and multivariable-IVW methods were the main analysis methods, and the other 15 pleiotropy-robust methods, weak IV-robust methods, and outliers-robust methods were used as auxiliary methods. RESULTS Maternal gestational age (early preterm birth, preterm birth, gestational duration, and post-term birth) had a causal relationship with 42 eye diseases. Four neonatal cytokines, Tumor Necrosis Factor-α(TNF-α), IL10, GROA, and CTACK, as well as four adult cytokines, CTACK, IL10, IL12p70 and IL6 are mediators in the causal relationships between early preterm birth and preterm birth in eight eye diseases. However, after adjusting for these mediators, a null direct causal effect of early preterm birth and preterm birth on eight eye diseases was found. In addition, there was no mediator in the causal relationship between gestational duration and post-term birth to eye diseases. CONCLUSION The effects of maternal gestational duration on offspring eye diseases through cytokines are long-term and life-course effects.
Collapse
Affiliation(s)
- Lei Hou
- Beijing International Center for Mathematical Research, Peking University, Beijing 100871, China
| | - Yunxia Li
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong 250022, China; Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong 250022, China
| | - Lili Kang
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong 250022, China; Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong 250022, China
| | - Xiaoying Li
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong 250022, China; Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong 250022, China.
| | - Hongkai Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China; Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250000, China.
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China; Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250000, China.
| |
Collapse
|
6
|
Ștefan G, Cinca S, Chiriac C, Zugravu A, Stancu S. Multiple Myeloma and Kidney Impairment at Diagnosis: A Nephrological Perspective from an Eastern European Country. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1326. [PMID: 37512137 PMCID: PMC10384718 DOI: 10.3390/medicina59071326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: The clinical presentation and survival factors in patients with myeloma-related kidney impairment (MRKI) at diagnosis remain a topic of ongoing research, given the complex interplay between nephrology and hematology. To date, no studies have specifically reported outcomes for these patients in Eastern Europe. Materials and Methods: We conducted a retrospective, unicentric study of consecutive newly diagnosed patients with MRKI in our tertiary nephrology service in Romania between 2015 and 2020; follow-up extended until 1 September 2022, covering a study period of 90 months. Results: We identified 89 consecutive patients with MRKI (median age 66 years, 38% male, median eGFR 5 mL/min). The majority of patients had arterial hypertension (71%) and systemic atherosclerosis (58%), and the most frequent clinical features at presentation were asthenia (75%) and bone pain (51%). Light-chain-restricted myeloma was the most common type (55%), with kappa free light chain being more frequent (53%). Among the patients, 81% presented with acute kidney injury (AKI), and 38% required hemodialysis at diagnosis. During the study period, 65% of the patients died, and hypoalbuminemia and the need for hemodialysis at diagnosis were significantly associated with mortality in multivariate analysis. Conclusions: Patients with MRKI who present to the nephrologist more frequently exhibit light chain restriction and most often present with AKI, with one-third requiring hemodialysis at diagnosis. Moreover, hypoalbuminemia and the initiation of hemodialysis at diagnosis were significantly associated with increased mortality.
Collapse
Affiliation(s)
- Gabriel Ștefan
- Faculty of Medicine, "Carol Davila" University of Medcine and Pharmacy, 050474 Bucharest, Romania
- Nephrology Department, "Dr. Carol Davila" Teaching Hospital of Nephrology, 010731 Bucharest, Romania
| | - Simona Cinca
- Faculty of Medicine, "Carol Davila" University of Medcine and Pharmacy, 050474 Bucharest, Romania
- Nephrology Department, "Dr. Carol Davila" Teaching Hospital of Nephrology, 010731 Bucharest, Romania
| | - Corina Chiriac
- Faculty of Medicine, "Carol Davila" University of Medcine and Pharmacy, 050474 Bucharest, Romania
- Nephrology Department, "Dr. Carol Davila" Teaching Hospital of Nephrology, 010731 Bucharest, Romania
| | - Adrian Zugravu
- Faculty of Medicine, "Carol Davila" University of Medcine and Pharmacy, 050474 Bucharest, Romania
- Nephrology Department, "Dr. Carol Davila" Teaching Hospital of Nephrology, 010731 Bucharest, Romania
| | - Simona Stancu
- Faculty of Medicine, "Carol Davila" University of Medcine and Pharmacy, 050474 Bucharest, Romania
- Nephrology Department, "Dr. Carol Davila" Teaching Hospital of Nephrology, 010731 Bucharest, Romania
| |
Collapse
|
7
|
CD8 + T Cell Senescence: Lights and Shadows in Viral Infections, Autoimmune Disorders and Cancer. Int J Mol Sci 2022; 23:ijms23063374. [PMID: 35328795 PMCID: PMC8955595 DOI: 10.3390/ijms23063374] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
CD8+ T lymphocytes are a heterogeneous class of cells that play a crucial role in the adaptive immune response against pathogens and cancer. During their lifetime, they acquire cytotoxic functions to ensure the clearance of infected or transformed cells and, in addition, they turn into memory lymphocytes, thus providing a long-term protection. During ageing, the thymic involution causes a reduction of circulating T cells and an enrichment of memory cells, partially explaining the lowering of the response towards novel antigens with implications in vaccine efficacy. Moreover, the persistent stimulation by several antigens throughout life favors the switching of CD8+ T cells towards a senescent phenotype contributing to a low-grade inflammation that is a major component of several ageing-related diseases. In genetically predisposed young people, an immunological stress caused by viral infections (e.g., HIV, CMV, SARS-CoV-2), autoimmune disorders or tumor microenvironment (TME) could mimic the ageing status with the consequent acceleration of T cell senescence. This, in turn, exacerbates the inflamed conditions with dramatic effects on the clinical progression of the disease. A better characterization of the phenotype as well as the functions of senescent CD8+ T cells can be pivotal to prevent age-related diseases, to improve vaccine strategies and, possibly, immunotherapies in autoimmune diseases and cancer.
Collapse
|
8
|
Visram A, Kourelis TV. Aging-associated immune system changes in multiple myeloma: The dark side of the moon. Cancer Treat Res Commun 2021; 29:100494. [PMID: 34837796 DOI: 10.1016/j.ctarc.2021.100494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 11/18/2021] [Indexed: 02/08/2023]
Abstract
Multiple myeloma (MM) is a disease of the elderly. Changes that occur in the immune system with aging, also known as immunosenescence, have been associated with decreased tumor immunosurveillance and are thought to contribute to the development of MM and other cancers in the elderly. Once MM establishes itself in the bone marrow, immunosenescence related changes have been observed in the immune tumor microenvironment (iTME) and are driven by the malignant cells. The efficacy of novel immunotherapies used to treat MM has been blunted by detrimental iTME changes that occur at later disease stages and are, to some extent, driven by prior therapies. In this review, we discuss general changes that occur in the immune system with aging as well as our current knowledge of immunosenescence in MM. We discuss the differences and overlap between T cell senescence and exhaustion as well as potential methods to prevent or reverse immunosenescence. We focus predominantly on T cell immunosenescence which has been better evaluated in this disease and is more pertinent to novel MM immunotherapies. Our lack of understanding of the drivers of immunosenescence at each stage of the disease, from precursor stages to heavily pretreated MM, represents a major barrier to improving the efficacy of novel and existing therapies.
Collapse
Affiliation(s)
- Alissa Visram
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN United States; Department of Medicine, Division of Hematology, University of Ottawa, Ottawa Hospital Research Institute, Ontario, Canada
| | - Taxiarchis V Kourelis
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN United States.
| |
Collapse
|
9
|
A Comprehensive Review of the Genomics of Multiple Myeloma: Evolutionary Trajectories, Gene Expression Profiling, and Emerging Therapeutics. Cells 2021; 10:cells10081961. [PMID: 34440730 PMCID: PMC8391934 DOI: 10.3390/cells10081961] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/21/2021] [Accepted: 07/30/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a blood cancer characterized by the accumulation of malignant monoclonal plasma cells in the bone marrow. It develops through a series of premalignant plasma cell dyscrasia stages, most notable of which is the Monoclonal Gammopathy of Undetermined Significance (MGUS). Significant advances have been achieved in uncovering the genomic aberrancies underlying the pathogenesis of MGUS-MM. In this review, we discuss in-depth the genomic evolution of MM and focus on the prognostic implications of the accompanied molecular and cytogenetic aberrations. We also dive into the latest investigatory techniques used for the diagnoses and risk stratification of MM patients.
Collapse
|
10
|
Signaling Pathway Mediating Myeloma Cell Growth and Survival. Cancers (Basel) 2021; 13:cancers13020216. [PMID: 33435632 PMCID: PMC7827005 DOI: 10.3390/cancers13020216] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The bone marrow (BM) microenvironment plays a crucial role in pathogenesis of multiple myeloma (MM), and delineation of the intracellular signaling pathways activated in the BM microenvironment in MM cells is essential to develop novel therapeutic strategies to improve patient outcome. Abstract The multiple myeloma (MM) bone marrow (BM) microenvironment consists of different types of accessory cells. Both soluble factors (i.e., cytokines) secreted from these cells and adhesion of MM cells to these cells play crucial roles in activation of intracellular signaling pathways mediating MM cell growth, survival, migration, and drug resistance. Importantly, there is crosstalk between the signaling pathways, increasing the complexity of signal transduction networks in MM cells in the BM microenvironment, highlighting the requirement for combination treatment strategies to blocking multiple signaling pathways.
Collapse
|
11
|
Kang B, Park H, Kim B. Anticancer Activity and Underlying Mechanism of Phytochemicals against Multiple Myeloma. Int J Mol Sci 2019; 20:E2302. [PMID: 31075954 PMCID: PMC6539572 DOI: 10.3390/ijms20092302] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/03/2019] [Accepted: 05/04/2019] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM)-a common hematologic malignancy of plasma cells-accounts for substantial mortality and morbidity rates. Due to the advent of novel therapies such as immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and monoclonal antibodies (mAbs), response rates were increased and free survival and overall survival have been elevated. However, adverse events including toxicity, neuropathy or continuous relapse are still problems. Thus, development of novel drugs which have less side effects and more effective is needed. This review aims to recapitulate the pharmacologic anti-MM mechanisms of various phytochemicals, elucidating their molecular targets. Keywords related to MM and natural products were searched in PUBMED/MEDLINE. Phytochemicals have been reported to display a variety of anti-MM activities, including apoptosis, cell cycle arrest, antiangiogenesis, and miRNA modulation. Some phytochemicals sensitize the conventional therapies such as dexamethasone. Also, there are clinical trials with phytochemicals such as agaricus, curcumin, and Neovastat regarding MM treatment. Taken together, this review elucidated and categorized the evidences that natural products and their bioactive compounds could be potent drugs in treating MM.
Collapse
Affiliation(s)
- Beomku Kang
- College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea.
| | - Hyunmin Park
- Department of Pathology, College of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea.
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea.
- Department of Pathology, College of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea.
| |
Collapse
|
12
|
BF211, a derivative of bufalin, enhances the cytocidal effects in multiple myeloma cells by inhibiting the IL-6/JAK2/STAT3 pathway. Int Immunopharmacol 2018; 64:24-32. [DOI: 10.1016/j.intimp.2018.08.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/01/2018] [Accepted: 08/16/2018] [Indexed: 01/05/2023]
|
13
|
Nageshwari B, Merugu R. Effect of levamisole on expression of CD138 and interleukin-6 in human multiple myeloma cell lines. Indian J Cancer 2018; 54:566-571. [PMID: 29798960 DOI: 10.4103/ijc.ijc_349_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is a B-cell malignancy accounting for 0.8% of all cancer deaths globally. This malignancy is characterized by lytic bone disease renal insufficiency, anemia, hypercalcemia, and immunodeficiency. The myeloma cells have enhanced expression of CD138. CD138 is a transmembrane heparin sulfate glycoprotein expressed on different types of adherent and nonadherent cells.CD138 is used as a standard marker for identification of tumor cells. AIMS AND OBJECTIVES Despite introduction of many therapeutic agents, the management of multiple myeloma (MM) remains a challenge and search for new therapeutic agents is in progress. In this study, we attempted to evaluate the effect of an alkaline phosphatase inhibitor, levamisole on expression of CD138, and level of interleukin-6 (IL-6) in human MM cell lines RPMI 8226 and U266 B1. MATERIAL AND METHODS U266B1 and RPMI 8226 cell lines were obtained from the National Centre for Cell Sciences, Pune. Alkaline phosphatase assay, Interleukin-6 assay and CD138 expression on myeloma cells by flow cytometry were investigated when the cells were exposed to Levamisole. RESULTS Levamisole-mediated growth inhibition of myeloma cells in vitro is associated with a loss of CD138 and increased IL-6 secretion. The increased secretion of IL-6 by myeloma cells could be an attempt to protect themselves from apoptosis. CONCLUSION Levamisole inhibited CD138 expression and affected the levels of IL-6 in a dose-dependent manner. The results of the present study add new dimension to levamisole's mode of action as inhibitor of CD138 and IL-6 and as an antiapoptotic agent.
Collapse
Affiliation(s)
- B Nageshwari
- Department of Biotechnology, Government College Autonomous, Rajamahendravaram, Andhra Pradesh, India
| | - Ramchander Merugu
- Department of Biochemistry, Mahatma Gandhi University, Nalgonda, Telangana, India
| |
Collapse
|
14
|
Burwick N, Sharma S. Glucocorticoids in multiple myeloma: past, present, and future. Ann Hematol 2018; 98:19-28. [PMID: 30073393 DOI: 10.1007/s00277-018-3465-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Glucocorticoids are a backbone of treatment for multiple myeloma in both the upfront and relapsed/refractory setting. While glucocorticoids have single agent activity in multiple myeloma, in the modern era, they are paired with novel agents to induce high clinical response rates. On the other hand, toxicities of steroid therapy limit high dose delivery and impact patient quality of life. We provide a history of steroid use in multiple myeloma with the aim to understand how steroids have emerged and persisted in the treatment of multiple myeloma. We review mechanisms of glucocorticoid sensitivity and resistance and highlight potential future directions to evaluate steroid responsiveness. Further research in this area will aid in optimizing steroid utilization and help determine when glucocorticoid therapy may no longer benefit patients.
Collapse
Affiliation(s)
- Nicholas Burwick
- VA Puget Sound Health Care System, Seattle, WA, USA. .,Department of Medicine, University of Washington, 1705 NE Pacific St, M/S 358280, Seattle, WA, 98195, USA.
| | | |
Collapse
|
15
|
López-Relaño J, Martín-Adrados B, Real-Arévalo I, Lozano-Bartolomé J, Abós B, Sánchez-Ramón S, Alonso B, Gómez Del Moral M, Martínez-Naves E. Monocyte-Derived Dendritic Cells Differentiated in the Presence of Lenalidomide Display a Semi-Mature Phenotype, Enhanced Phagocytic Capacity, and Th1 Polarization Capability. Front Immunol 2018; 9:1328. [PMID: 29951065 PMCID: PMC6008535 DOI: 10.3389/fimmu.2018.01328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 05/28/2018] [Indexed: 11/16/2022] Open
Abstract
Lenalidomide is an analog of thalidomide, with potent anticancer activity demonstrated in several hematological malignancies. It has immunomodulatory properties, being able to enhance the activation of different types of immune cells, which results in antitumor activities. Dendritic cells (DCs) are pivotal in the immune response, and different immunotherapeutic approaches targeting these cells are being developed. Since little is known about the effect of lenalidomide on DCs, the goal of the present work was to investigate the phenotype and function of human monocyte-derived DCs differentiated in the presence of lenalidomide (L-DCs). Our results showed that L-DCs display a unique phenotype, with increased cell surface expression of some maturation markers such as CD1d, CD83, CD86, and HLA-DR. This phenotype correlates with a lower expression of the E3 ubiquitin-ligase MARCH-I in L-DCs, upregulating the cell surface expression of CD86 and HLA-DR. In addition, immature L-DCs express higher amounts of DC-SIGN on the cell surface than control immature DCs. After LPS stimulation, production of IL-6 and TNF-α was severely decreased, whereas IL-12 and IL-10 secretion was dramatically upregulated in L-DCs, compared to that in the controls. Functionally, L-DCs are more effectively recognized by NKT cells in cytotoxicity experiments. Furthermore, L-DCs display higher opsonin-independent antigen uptake capability than control DCs. Mixed lymphocyte reaction experiments showed that L-DCs could stimulate naïve CD4 T-cells, polarizing them toward a predominant Th1 phenotype. In summary, DCs derived from monocytes in the presence of lenalidomide present a semi-mature phenotype, increased phagocytic capacity, reduced production of proinflammatory cytokines, and the ability to polarize T-cells toward predominant Th1-type responses; these are qualities that might be useful in the development of new immunotherapeutic treatments.
Collapse
Affiliation(s)
- Juan López-Relaño
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Beatriz Martín-Adrados
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Irene Real-Arévalo
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Javier Lozano-Bartolomé
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Beatriz Abós
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | | | | | - Manuel Gómez Del Moral
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain.,Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Eduardo Martínez-Naves
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| |
Collapse
|
16
|
Rozic G, Paukov L, Jakubikova J, Ben-Shushan D, Duek A, Leiba A, Avigdor A, Nagler A, Leiba M. The novel compound STK405759 is a microtubule-targeting agent with potent and selective cytotoxicity against multiple myeloma in vitro and in vivo. Oncotarget 2018; 7:62572-62584. [PMID: 27613836 PMCID: PMC5308747 DOI: 10.18632/oncotarget.11539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/09/2016] [Indexed: 12/31/2022] Open
Abstract
Despite advances in treatment, multiple myeloma (MM) remains incurable. Here we propose the use of STK405759, a novel microtubule targeting agent (MTA) and member of the furan metotica family for MM therapy. STK405759 inhibited tubulin polymerization in a cell-free system and in myeloma cells. This molecule had potent cytotoxic activity against several MM cell lines and patient-derived MM cells. Moreover, STK405759 demonstrated cytotoxicity against drug-resistant myeloma cells that overexpressed the P-glycoprotein drug-efflux pump. STK405759 was not cytotoxic to peripheral blood mononuclear cells, including activated B and T lymphocytes. This compound caused mitotic arrest and apoptosis of myeloma cells characterized by cleavage of poly (ADP-ribose) polymerase-1 and caspase-8, as well as decreased protein expression of mcl-1. The combination of STK405759 with bortezomib, lenalidomide or dexamethasone had synergistic cytotoxic activity. In in vivo studies, STK405759-treated mice had significantly decreased MM tumor burden and prolonged survival compared to vehicle treated- mice. These results provide a rationale for further evaluation of STK405759 as monotherapy or part of combination therapy for treating patients with MM.
Collapse
Affiliation(s)
- Gabriela Rozic
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Lena Paukov
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Jana Jakubikova
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dikla Ben-Shushan
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Adrian Duek
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Adi Leiba
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Medical Education, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Abraham Avigdor
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Nagler
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Merav Leiba
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Jakubikova J, Cholujova D, Hideshima T, Gronesova P, Soltysova A, Harada T, Joo J, Kong SY, Szalat RE, Richardson PG, Munshi NC, Dorfman DM, Anderson KC. A novel 3D mesenchymal stem cell model of the multiple myeloma bone marrow niche: biologic and clinical applications. Oncotarget 2018; 7:77326-77341. [PMID: 27764795 PMCID: PMC5357212 DOI: 10.18632/oncotarget.12643] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/28/2016] [Indexed: 12/19/2022] Open
Abstract
Specific niches within the tumor bone marrow (BM) microenvironment afford a sanctuary for multiple myeloma (MM) clones due to stromal cell-tumor cell interactions, which confer survival advantage and drug resistance. Defining the sequelae of tumor cell interactions within the MM niches on an individualized basis may provide the rationale for personalized therapies. To mimic the MM niche, we here describe a new 3D co-culture ex-vivo model in which primary MM patient BM cells are co-cultured with mesenchymal stem cells (MSC) in a hydrogel 3D system. In the 3D model, MSC with conserved phenotype (CD73+CD90+CD105+) formed compact clusters with active fibrous connections, and retained lineage differentiation capacity. Extracellular matrix molecules, integrins, and niche related molecules including N-cadherin and CXCL12 are expressed in 3D MSC model. Furthermore, activation of osteogenesis (MMP13, SPP1, ADAMTS4, and MGP genes) and osteoblastogenic differentiation was confirmed in 3D MSC model. Co-culture of patient-derived BM mononuclear cells with either autologous or allogeneic MSC in 3D model increased proliferation of MM cells, CXCR4 expression, and SP cells. We carried out immune profiling to show that distribution of immune cell subsets was similar in 3D and 2D MSC model systems. Importantly, resistance to novel agents (IMiDs, bortezomib, carfilzomib) and conventional agents (doxorubicin, dexamethasone, melphalan) was observed in 3D MSC system, reflective of clinical resistance. This 3D MSC model may therefore allow for studies of MM pathogenesis and drug resistance within the BM niche. Importantly, ongoing prospective trials are evaluating its utility to inform personalized targeted and immune therapy in MM.
Collapse
Affiliation(s)
- Jana Jakubikova
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Danka Cholujova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Teru Hideshima
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paulina Gronesova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Andrea Soltysova
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Takeshi Harada
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jungnam Joo
- Biometric Research Branch, Division of Cancer Epidemiology and Prevention, Research Institute & Hospital, National Cancer Center, Goyang-si Gyeonggi-do, South Korea
| | - Sun-Young Kong
- Department of Laboratory Medicine and Translational Epidemiology Branch, Research Institute & Hospital, National Cancer Center, Goyang-si Gyeonggi-do, South Korea
| | - Raphael E Szalat
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paul G Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nikhil C Munshi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Microenvironment drug resistance in multiple myeloma: emerging new players. Oncotarget 2018; 7:60698-60711. [PMID: 27474171 PMCID: PMC5312413 DOI: 10.18632/oncotarget.10849] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/11/2016] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) drug resistance (DR) is a multistep transformation process based on a powerful interplay between bone marrow stromal cells and MM cells that allows the latter to escape anti-myeloma therapies. Here we present an overview of the role of the bone marrow microenvironment in both soluble factors-mediated drug resistance (SFM-DR) and cell adhesion-mediated drug resistance (CAM-DR), focusing on the role of new players, namely miRNAs, exosomes and cancer-associated fibroblasts.
Collapse
|
19
|
Bar-Natan M, Stroopinsky D, Luptakova K, Coll MD, Apel A, Rajabi H, Pyzer AR, Palmer K, Reagan MR, Nahas MR, Leaf RK, Jain S, Arnason J, Ghobrial IM, Anderson KC, Kufe D, Rosenblatt J, Avigan D. Bone marrow stroma protects myeloma cells from cytotoxic damage via induction of the oncoprotein MUC1. Br J Haematol 2017; 176:929-938. [PMID: 28107546 PMCID: PMC5800979 DOI: 10.1111/bjh.14493] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/10/2016] [Indexed: 01/19/2023]
Abstract
Multiple myeloma (MM) is a lethal haematological malignancy that arises in the context of a tumour microenvironment that promotes resistance to apoptosis and immune escape. In the present study, we demonstrate that co-culture of MM cells with stromal cells results in increased resistance to cytotoxic and biological agents as manifested by decreased rates of cell death following exposure to alkylating agents and the proteosome inhibitor, bortezomib. To identify the mechanism of increased resistance, we examined the effect of the co-culture of MM cells with stroma cells, on expression of the MUC1 oncogene, known to confer tumour cells with resistance to apoptosis and necrosis. Co-culture of stroma with MM cells resulted in increased MUC1 expression by tumour cells. The effect of stromal cell co-culture on MUC1 expression was not dependent on cell contact and was therefore thought to be due to soluble factors secreted by the stromal cells into the microenvironment. We demonstrated that MUC1 expression was mediated by interleukin-6 and subsequent up-regulation of the JAK-STAT pathway. Interestingly, the effect of stromal cell co-culture on tumour resistance was partially reversed by silencing of MUC1 in MM cells, consistent with the potential role of MUC1 in mediating resistance to cytotoxic-based therapies.
Collapse
Affiliation(s)
| | | | | | - Maxwell D. Coll
- Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Arie Apel
- Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Hasan Rajabi
- Dana Farber Cancer Institute, Harvard Medical School and
| | | | - Kristen Palmer
- Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Michaela R. Reagan
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Myrna R. Nahas
- Beth Israel Deaconess Medical Center, Harvard Medical School
| | | | - Salvia Jain
- Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Jon Arnason
- Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Irene M. Ghobrial
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Donald Kufe
- Dana Farber Cancer Institute, Harvard Medical School and
| | | | - David Avigan
- Beth Israel Deaconess Medical Center, Harvard Medical School
| |
Collapse
|
20
|
Tazawa S, Shiozawa E, Homma M, Arai N, Kabasawa N, Kawaguchi Y, Fujiwara S, Okino K, Kobayashi K, Yamochi T, Tate G, Nakamaki T, Takimoto M. CD200 Expression on Plasma Cell Myeloma Cells is Associated with the Efficacies of Bortezomib, Lenalidomide and Thalidomide. J Clin Exp Hematop 2016; 55:121-6. [PMID: 26763359 DOI: 10.3960/jslrt.55.121] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Plasma cell myeloma (PCM) is a devastating disease with a highly heterogeneous outcome, with survival ranging from a few months to longer than 10 years. Treatment of multiple myeloma has changed markedly in the past decade due to the development of new drugs such as bortezomib, lenalidomide and thalidomide, which have greatly improved the outcome of PCM. The clinical and prognostic value of immunophenotyping in PCM remains questionable. The aim of this study was to determine the diagnostic and prognostic significance of CD200 expression in newly diagnosed PCM. We retrospectively reviewed the records of 107 patients newly diagnosed with PCM at Showa University Hospital between January 2004 and September 2013. Expression of CD200 was studied by immunohistochemistry. Clinical and pathological parameters were compared between CD200-positive and CD200-negative cases. CD200-positive PCM cases had lower serum albumin (p = 0.0001) compared to those without CD200 expression. Our results showed no significant difference in median overall survival between patients with CD200-positive and CD200-negative PCM. However, there was a strong correlation between CD200 expression and serum albumin level. In the CD200-negative group, median overall survival was significantly longer in patients who received new drug treatment. These findings suggest that CD200 expression is a useful marker for evaluation of the severity of PCM and that lack of CD200 expression may improve the sensitivity of PCM to therapy with new drugs.
Collapse
Affiliation(s)
- Sakiko Tazawa
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Akl MR, Nagpal P, Ayoub NM, Tai B, Prabhu SA, Capac CM, Gliksman M, Goy A, Suh KS. Molecular and clinical significance of fibroblast growth factor 2 (FGF2 /bFGF) in malignancies of solid and hematological cancers for personalized therapies. Oncotarget 2016; 7:44735-44762. [PMID: 27007053 PMCID: PMC5190132 DOI: 10.18632/oncotarget.8203] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/10/2016] [Indexed: 12/30/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling is essential for normal and cancer biology. Mammalian FGF family members participate in multiple signaling pathways by binding to heparan sulfate and FGF receptors (FGFR) with varying affinities. FGF2 is the prototype member of the FGF family and interacts with its receptor to mediate receptor dimerization, phosphorylation, and activation of signaling pathways, such as Ras-MAPK and PI3K pathways. Excessive mitogenic signaling through the FGF/FGFR axis may induce carcinogenic effects by promoting cancer progression and increasing the angiogenic potential, which can lead to metastatic tumor phenotypes. Dysregulated FGF/FGFR signaling is associated with aggressive cancer phenotypes, enhanced chemotherapy resistance and poor clinical outcomes. In vitro experimental settings have indicated that extracellular FGF2 affects proliferation, drug sensitivity, and apoptosis of cancer cells. Therapeutically targeting FGF2 and FGFR has been extensively assessed in multiple preclinical studies and numerous drugs and treatment options have been tested in clinical trials. Diagnostic assays are used to quantify FGF2, FGFRs, and downstream signaling molecules to better select a target patient population for higher efficacy of cancer therapies. This review focuses on the prognostic significance of FGF2 in cancer with emphasis on therapeutic intervention strategies for solid and hematological malignancies.
Collapse
Affiliation(s)
- Mohamed R. Akl
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Poonam Nagpal
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Betty Tai
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Sathyen A. Prabhu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Catherine M. Capac
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Matthew Gliksman
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Lymphoma Division, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K. Stephen Suh
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
22
|
Effects of IL-8 Up-Regulation on Cell Survival and Osteoclastogenesis in Multiple Myeloma. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2171-2182. [PMID: 27301357 DOI: 10.1016/j.ajpath.2016.04.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/14/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023]
Abstract
IL-8 promotes cancer cell growth, survival, angiogenesis, and metastasis in several tumors. Herein, we investigated the sources of IL-8 production in multiple myeloma (MM) and its potential roles in MM pathogenesis. We found that bone marrow cells from patients with MM secreted higher amounts of IL-8 than healthy donors. IL-8 production was detected in cultures of CD138(+) plasma cells and CD138(-) cells isolated from bone marrows of MM patients, and in three of seven human myeloma cell lines (HMCLs) analyzed. Interactions between MM and stromal cells increased IL-8 secretion by stromal cells through cell-cell adhesion and soluble factors. Interestingly, IL8 expression also increased in HMCLs, stromal cells, and osteoclasts after treatment with the antimyeloma drugs melphalan and bortezomib. In fact, the effect of bortezomib on IL-8 production was higher than that exerted by stromal-MM cell interactions. Addition of exogenous IL-8 did not affect growth of HMCLs, although it protected cells from death induced by serum starvation through a caspase-independent mechanism. Furthermore, IL-8 induced by stromal-MM cell interactions strongly contributed to osteoclast formation in vitro, because osteoclastogenesis was markedly reduced by IL-8-specific neutralizing antibodies. In conclusion, our results implicate IL-8 in myeloma bone disease and point to the potential utility of an anti-IL-8 therapy to prevent unwanted effects of IL-8 up-regulation on survival, angiogenesis, and osteolysis in MM.
Collapse
|
23
|
Shah JJ, Feng L, Thomas SK, Berkova Z, Weber DM, Wang M, Qazilbash MH, Champlin RE, Mendoza TR, Cleeland C, Orlowski RZ. Siltuximab (CNTO 328) with lenalidomide, bortezomib and dexamethasone in newly-diagnosed, previously untreated multiple myeloma: an open-label phase I trial. Blood Cancer J 2016; 6:e396. [PMID: 26871714 PMCID: PMC4771967 DOI: 10.1038/bcj.2016.4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/04/2016] [Indexed: 01/01/2023] Open
Abstract
The safety and efficacy of siltuximab (CNTO 328) was tested in combination with lenalidomide, bortezomib and dexamethasone (RVD) in patients with newly-diagnosed, previously untreated symptomatic multiple myeloma. Fourteen patients were enrolled in the study, eleven of whom qualified to receive therapy. A majority of patients (81.8%) completed the minimal number or more of the four required cycles, while two patients completed only three cycles. The maximum tolerated dose (MTD) of siltuximab with RVD was dose level -1 (siltuximab: 8.3 mg/kg; bortezomib: 1.3 mg/m(2); lenalidomide: 25 mg; dexamethasone: 20 mg). Serious adverse events were grade 3 pneumonia and grade 4 thrombocytopenia, and no deaths occurred during the study or with follow-up (median follow-up 28.1 months). An overall response rate, after 3-4 cycles of therapy, of 90.9% (95% confidence interval (CI): 58.7%, 99.8%) (9.1% complete response (95% CI: 0.2%, 41.3%), 45.5% very good partial response (95% CI: 16.7%, 76.6%) and 36.4% partial response (95% CI: 10.9%, 69.2%)) was seen. Two patients withdrew consent, and nine patients (81.8%) opted for autologous stem cell transplantation.
Collapse
Affiliation(s)
- J J Shah
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S K Thomas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Z Berkova
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D M Weber
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M H Qazilbash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R E Champlin
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T R Mendoza
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C Cleeland
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Mechanisms of Drug Resistance in Relapse and Refractory Multiple Myeloma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:341430. [PMID: 26649299 PMCID: PMC4663284 DOI: 10.1155/2015/341430] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/24/2015] [Accepted: 10/21/2015] [Indexed: 12/11/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy that remains incurable because most patients eventually relapse or become refractory to current treatments. Although the treatments have improved, the major problem in MM is resistance to therapy. Clonal evolution of MM cells and bone marrow microenvironment changes contribute to drug resistance. Some mechanisms affect both MM cells and microenvironment, including the up- and downregulation of microRNAs and programmed death factor 1 (PD-1)/PD-L1 interaction. Here, we review the pathogenesis of MM cells and bone marrow microenvironment and highlight possible drug resistance mechanisms. We also review a potential molecular targeting treatment and immunotherapy for patients with refractory or relapse MM.
Collapse
|
25
|
Thompson PA, Khatami M, Baglole CJ, Sun J, Harris S, Moon EY, Al-Mulla F, Al-Temaimi R, Brown D, Colacci A, Mondello C, Raju J, Ryan E, Woodrick J, Scovassi I, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Hamid RA, Lowe L, Guarnieri T, Bisson WH. Environmental immune disruptors, inflammation and cancer risk. Carcinogenesis 2015; 36 Suppl 1:S232-S253. [PMID: 26106141 PMCID: PMC4492068 DOI: 10.1093/carcin/bgv038] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 01/09/2015] [Accepted: 01/14/2015] [Indexed: 12/16/2022] Open
Abstract
An emerging area in environmental toxicology is the role that chemicals and chemical mixtures have on the cells of the human immune system. This is an important area of research that has been most widely pursued in relation to autoimmune diseases and allergy/asthma as opposed to cancer causation. This is despite the well-recognized role that innate and adaptive immunity play as essential factors in tumorigenesis. Here, we review the role that the innate immune cells of inflammatory responses play in tumorigenesis. Focus is placed on the molecules and pathways that have been mechanistically linked with tumor-associated inflammation. Within the context of chemically induced disturbances in immune function as co-factors in carcinogenesis, the evidence linking environmental toxicant exposures with perturbation in the balance between pro- and anti-inflammatory responses is reviewed. Reported effects of bisphenol A, atrazine, phthalates and other common toxicants on molecular and cellular targets involved in tumor-associated inflammation (e.g. cyclooxygenase/prostaglandin E2, nuclear factor kappa B, nitric oxide synthesis, cytokines and chemokines) are presented as example chemically mediated target molecule perturbations relevant to cancer. Commentary on areas of additional research including the need for innovation and integration of systems biology approaches to the study of environmental exposures and cancer causation are presented.
Collapse
Affiliation(s)
- Patricia A. Thompson
- *To whom correspondence should be addressed. Tel: +1 631 444 6818; Fax: +1 631 444 3424;
| | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), NIH, Bethesda, MD 20817, USA
| | - Carolyn J. Baglole
- Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, IL 60612, USA
| | - Shelley Harris
- Prevention and Cancer Control, Cancer Care Ontario, 620 University Avenue, Toronto, Ontario M5G 2L3, Canada
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of South Korea
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Dustin Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Chiara Mondello
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Ivana Scovassi
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | - Roslida A. Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra, Malaysia, Serdang, Selangor 43400, Malaysia
| | - Leroy Lowe
- Getting to Know Cancer, Room 229A, 36 Arthur St, Truro, Nova Scotia B2N 1X5, Canada
| | - Tiziana Guarnieri
- Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy
- Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, Via Massarenti, 9, 40126 Bologna, Italy,
- National Institute of Biostructures and Biosystems, Viale Medaglie d’ Oro, 305, 00136 Roma, Italy and
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
26
|
Béguelin W, Sawh S, Chambwe N, Chan FC, Jiang Y, Choo JW, Scott DW, Chalmers A, Geng H, Tsikitas L, Tam W, Bhagat G, Gascoyne RD, Shaknovich R. IL10 receptor is a novel therapeutic target in DLBCLs. Leukemia 2015; 29:1684-94. [PMID: 25733167 DOI: 10.1038/leu.2015.57] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 12/30/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a biologically and clinically heterogeneous disease with marked genomic instability and variable response to conventional R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone) chemotherapy. More clinically aggressive cases of DLBCLs have high level of circulating interleukin 10 (IL10) cytokine and evidence of activated intracellular STAT3 (signal transducer and activator of transcription 3) signaling. We investigated the role of IL10 and its surface receptor in supporting the neoplastic phenotype of DLBCLs. We determined that IL10RA gene is amplified in 21% and IL10RB gene in 10% of primary DLBCLs. Gene expression of IL10, IL10RA and IL10RB was markedly elevated in DLBCLs. We hypothesized that DLBCLs depend for their proliferation and survival on IL10-STAT3 signaling and that blocking the IL10 receptor (IL10R) would induce cell death. We used anti-IL10R blocking antibody, which resulted in a dose-dependent cell death in all tested activated B-cell-like subtype of DLBCL cell lines and primary DLBCLs. Response of germinal center B-cell-like subtype of DLBCL cell lines to anti-IL10R antibody varied from sensitive to resistant. Cells underwent cell cycle arrest, followed by induction of apoptosis. Cell death depended on inhibition of STAT3 and, to a lesser extent, STAT1 signaling. Anti-IL10R treatment resulted in interruption of IL10-IL10R autostimulatory loop. We thus propose that IL10R is a novel therapeutic target in DLBCLs.
Collapse
Affiliation(s)
- W Béguelin
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA
| | - S Sawh
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA
| | - N Chambwe
- 1] The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA [2] Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA [3] Tri-Instituitional Training Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, NY, USA
| | - F C Chan
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Y Jiang
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA
| | - J-W Choo
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA
| | - D W Scott
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - A Chalmers
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA
| | - H Geng
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - L Tsikitas
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA
| | - W Tam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - G Bhagat
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - R D Gascoyne
- 1] Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC, Canada [2] Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - R Shaknovich
- 1] Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA [2] Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
27
|
Suzuki K, Ogura M, Abe Y, Suzuki T, Tobinai K, Ando K, Taniwaki M, Maruyama D, Kojima M, Kuroda J, Achira M, Iizuka K. Phase 1 study in Japan of siltuximab, an anti-IL-6 monoclonal antibody, in relapsed/refractory multiple myeloma. Int J Hematol 2015; 101:286-94. [PMID: 25655379 DOI: 10.1007/s12185-015-1743-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 01/08/2015] [Accepted: 01/15/2015] [Indexed: 01/30/2023]
Abstract
Siltuximab, a chimeric monoclonal antibody with high affinity and specificity for interleukin-6, has been shown to enhance anti-multiple myeloma activity of bortezomib and corticosteroid in vitro. We evaluated the safety, pharmacokinetics, immunogenicity, and antitumor effect of siltuximab in combination with bortezomib and dexamethasone in Japanese patients with relapsed or refractory multiple myeloma. This open-label, phase 1, dose-escalating study used two doses of siltuximab: 5.5 and 11.0 mg/kg (administered on day 1 of each 21-day cycle). In total, nine patients were treated. The most common grade 3/4 adverse events, lymphopenia (89 %) and thrombocytopenia (44 %), occurred in patients receiving both doses of siltuximab; however, no dose-limiting toxicities (DLTs) were observed. Following intravenous administration of siltuximab at 5.5 and 11.0 mg/kg, the maximum serum concentration and the area under the curve from 0 to 21 days and from 0 to infinity increased in an approximately dose-proportional manner. Mean half-life, total systemic clearance, and volume of distribution were similar at doses of 5.5 and 11.0 mg/kg. Across both doses, six of the nine patients had complete or partial response (22 and 44 %, respectively). In conclusion, as no DLT was observed, the recommended dose for this combination is 11.0 mg/kg once every 3 weeks. The study is registered at http://www.clinicaltrials.gov as NCT01309412.
Collapse
Affiliation(s)
- Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, Hiroo 4-1-22 Shibuya-ku, Tokyo, 1508935, Japan,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
The tumor microenvironment shapes hallmarks of mature B-cell malignancies. Oncogene 2015; 34:4673-82. [PMID: 25639873 DOI: 10.1038/onc.2014.403] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023]
Abstract
B-cell tumorigenesis results from a host of known and unknown genetic anomalies, including non-random translocations of genes that normally function as determinants of cell proliferation or cell survival to regions juxtaposed to active immunoglobulin heavy chain enhancer elements, chromosomal aneuploidy, somatic mutations that further affect oncogenic signaling and loss of heterozygosity of tumor-suppressor genes. However, it is critical to recognize that even in the setting of a genetic disease, the B-cell/plasma cell tumor microenvironment (TME) contributes significantly to malignant transformation and pathogenesis. Over a decade ago, we proposed the concept of cell adhesion-mediated drug resistance to delineate a form of TME-mediated drug resistance that protects hematopoietic tumor cells from the initial effect of diverse therapies. In the interim, it has been increasingly appreciated that TME also contributes to tumor initiation and progression through sustained growth/proliferation, self-renewal capacity, immune evasion, migration and invasion as well as resistance to cell death in a host of B-cell malignancies, including mantle cell lymphoma, diffuse large B-cell lymphoma, Waldenstroms macroglobulinemia, chronic lymphocytic leukemia and multiple myeloma. Within this review, we propose that TME and the tumor co-evolve as a consequence of bidirectional signaling networks. As such, TME represents an important target and should be considered integral to tumor progression and drug response.
Collapse
|
29
|
Abdi J, Chen G, Chang H. Drug resistance in multiple myeloma: latest findings and new concepts on molecular mechanisms. Oncotarget 2014; 4:2186-207. [PMID: 24327604 PMCID: PMC3926819 DOI: 10.18632/oncotarget.1497] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In the era of new and mostly effective therapeutic protocols, multiple myeloma still tends to be a hard-to-treat hematologic cancer. This hallmark of the disease is in fact a sequel to drug resistant phenotypes persisting initially or emerging in the course of treatment. Furthermore, the heterogeneous nature of multiple myeloma makes treating patients with the same drug challenging because finding a drugable oncogenic process common to all patients is not yet feasible, while our current knowledge of genetic/epigenetic basis of multiple myeloma pathogenesis is outstanding. Nonetheless, bone marrow microenvironment components are well known as playing critical roles in myeloma tumor cell survival and environment-mediated drug resistance happening most possibly in all myeloma patients. Generally speaking, however; real mechanisms underlying drug resistance in multiple myeloma are not completely understood. The present review will discuss the latest findings and concepts in this regard. It reviews the association of important chromosomal translocations, oncogenes (e.g. TP53) mutations and deranged signaling pathways (e.g. NFκB) with drug response in clinical and experimental investigations. It will also highlight how bone marrow microenvironment signals (Wnt, Notch) and myeloma cancer stem cells could contribute to drug resistance in multiple myeloma.
Collapse
Affiliation(s)
- Jahangir Abdi
- Dept. of Laboratory Medicine & Pathobiology, University of Toronto, Ontario, Canada
| | | | | |
Collapse
|
30
|
Park EJ, Lee SY, Lee GH, Kim DW, Kim Y, Cho MH, Kim JH. Sheet-type titania, but not P25, induced paraptosis accompanying apoptosis in murine alveolar macrophage cells. Toxicol Lett 2014; 230:69-79. [PMID: 25111187 DOI: 10.1016/j.toxlet.2014.07.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 02/08/2023]
Abstract
In this study, we identified the toxic effects of sheet-type titania (TNS), which are being developed as a material for UV-blocking glass, comparing with P25, a benchmark control for titania, in MH-S cells, a mouse alveolar macrophage cell line. After 24 h exposure, the TNS-exposed cells formed large vacuoles while the P25-exposed ones did not. The decreased levels of cell viability were similar between the P25 and TNS groups, but ATP production was clearly lower in cells exposed to the TNS. P25 decreased the expression of calnexin protein, an endoplasmic reticulum (ER) membrane marker, and increased the number of cells generating ROS in a dose dependent manner. Meanwhile, TNS dilated the ER and mitochondria and increased the secretion of NO and pro-inflammatory cytokines, but not of ROS. Subsequently, we studied the molecular response following TNS-induced vacuolization. TNS started to form vacuoles in the cytosol since 20 min after exposure, and the expression of the mitochondria function-related genes were down-regulated the most in the cells exposed for 1 h. After 24 h exposure, the number of apoptotic cells and the relative levels of BAX to Bcl-2 increased. The expression of SOD1 protein, but not of SOD2, also dose-dependently increased with an increase in caspase-8 activity. Additionally, the MAPK pathway was significantly activated, even though the expression of p-EGFR did not change significantly. Furthermore, the number of apoptotic cells increased rapidly with time and with the inhibition of vacuole formation. Taken together, we suggest that P25 and TNS may target different organelles. In addition, TNS, but not P25, induced paraptosis accompanied by apoptosis in MH-S cells, and the formation of the cytoplasmic vacuoles allowed delay apoptosis following TNS exposure.
Collapse
Affiliation(s)
- Eun-Jung Park
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Republic of Korea.
| | - Seung Yun Lee
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Republic of Korea
| | - Gwang-Hee Lee
- School of Civil, Environmental and Architectural Engineering, Korea University, Seoul 136-713, Republic of Korea
| | - Dong-Wan Kim
- School of Civil, Environmental and Architectural Engineering, Korea University, Seoul 136-713, Republic of Korea
| | - Younghun Kim
- Department of Chemical Engineering, Kwangwoon University, Seoul 139-701, Republic of Korea
| | - Myung-Haing Cho
- College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Republic of Korea.
| |
Collapse
|
31
|
Rosean TR, Tompkins VS, Tricot G, Holman CJ, Olivier AK, Zhan F, Janz S. Preclinical validation of interleukin 6 as a therapeutic target in multiple myeloma. Immunol Res 2014; 59:188-202. [PMID: 24845460 PMCID: PMC4209159 DOI: 10.1007/s12026-014-8528-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Studies on the biologic and molecular genetic underpinnings of multiple myeloma (MM) have identified the pleiotropic, pro-inflammatory cytokine, interleukin-6 (IL-6), as a factor crucial to the growth, proliferation and survival of myeloma cells. IL-6 is also a potent stimulator of osteoclastogenesis and a sculptor of the tumor microenvironment in the bone marrow of patients with myeloma. This knowledge has engendered considerable interest in targeting IL-6 for therapeutic purposes, using a variety of antibody- and small-molecule-based therapies. However, despite the early recognition of the importance of IL-6 for myeloma and the steady progress in our knowledge of IL-6 in normal and malignant development of plasma cells, additional efforts will be required to translate the promise of IL-6 as a target for new myeloma therapies into significant clinical benefits for patients with myeloma. This review summarizes published research on the role of IL-6 in myeloma development and describes ongoing efforts by the University of Iowa Myeloma Multidisciplinary Oncology Group to develop new approaches to the design and testing of IL-6-targeted therapies and preventions of MM.
Collapse
Affiliation(s)
- Timothy R Rosean
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Ghosh N, Tucker N, Zahurak M, Wozney J, Borrello I, Huff CA. Clarithromycin overcomes resistance to lenalidomide and dexamethasone in multiple myeloma. Am J Hematol 2014; 89:E116-20. [PMID: 24723438 DOI: 10.1002/ajh.23733] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 03/25/2014] [Accepted: 04/07/2014] [Indexed: 12/17/2022]
Abstract
The combination of clarithromycin, lenalidomide and dexamethasone (BiRd) has led to highly durable responses in newly diagnosed myeloma. However, the ability of clarithromycin to overcome resistance to lenalidomide and dexamethasone (Rd) is not known. To study this, we performed a retrospective analysis of 24 patients with myeloma for which clarithromycin was added to Rd at the time of progression on Rd. The median number of prior therapies was 3 (range 1-8). The best response was complete response (CR) in one (4.2%), very good partial response (VGPR) in one (4.2%) and partial response in eight (33.3%) patients. Ten patients, 41.7% (95% CI: 22.1, 63.4), achieved ≥PR. The median time to response was 4.4 months (range 1-13.6 months) and the median duration of response was 6.9 months (range 3-52.2 months). The clinical benefit rate (CR + VGPR + PR + MR) was 45.8% (95% CI 25.6, 67.2). The median progression-free survival was 4 months. Median overall survival was 25 months with a median follow-up of 27.5 months. The regimen was well tolerated and only 2 patients needed a clarithromycin dose reduction. Addition of clarithromycin to Rd can overcome resistance to Rd in a subset of patients and lead to durable clinical responses.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Levine Cancer Institute, Carolinas Healthcare System; Charlotte North Carolina
| | - Noah Tucker
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology; The Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Marianna Zahurak
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology; The Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Jocelyn Wozney
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology; The Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Ivan Borrello
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology; The Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Carol Ann Huff
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology; The Johns Hopkins University School of Medicine; Baltimore Maryland
| |
Collapse
|
33
|
Cho HY, Lee SW. TLR5 activation by flagellin induces doxorubicin resistance via interleukin-6 (IL-6) expression in two multiple myeloma cells. Cell Immunol 2014; 289:27-35. [DOI: 10.1016/j.cellimm.2014.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/28/2014] [Accepted: 03/13/2014] [Indexed: 02/08/2023]
|
34
|
Alexandrakis MG, Pappa CA, Kolovou A, Kyriakaki S, Vyzoukaki R, Devetzoglou M, Tsirakis G. Circulating levels of soluble Fas ligand reflect disease progression in multiple myeloma. Med Oncol 2014; 31:953. [DOI: 10.1007/s12032-014-0953-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 03/29/2014] [Indexed: 10/25/2022]
|
35
|
Nakajima S, Fujiwara T, Ohguchi H, Onishi Y, Kamata M, Okitsu Y, Fukuhara N, Ishizawa K, Harigae H. Induction of thymic stromal lymphopoietin in mesenchymal stem cells by interaction with myeloma cells. Leuk Lymphoma 2014; 55:2605-13. [DOI: 10.3109/10428194.2014.881478] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Shinji Nakajima
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Tohru Fujiwara
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
- Molecular Hematology/Oncology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Hiroto Ohguchi
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Yasushi Onishi
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Mayumi Kamata
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Yoko Okitsu
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Noriko Fukuhara
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Kenichi Ishizawa
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
- Molecular Hematology/Oncology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine,
Sendai, Japan
- Molecular Hematology/Oncology, Tohoku University Graduate School of Medicine,
Sendai, Japan
| |
Collapse
|
36
|
|
37
|
Semeraro M, Vacchelli E, Eggermont A, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Lenalidomide-based immunochemotherapy. Oncoimmunology 2013; 2:e26494. [PMID: 24482747 PMCID: PMC3897503 DOI: 10.4161/onci.26494] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 09/14/2013] [Indexed: 12/19/2022] Open
Abstract
Lenalidomide is a synthetic derivative of thalidomide currently approved by the US Food and Drug Administration for use in patients affected by multiple myeloma (in combination with dexamethasone) and low or intermediate-1 risk myelodysplastic syndromes that harbor 5q cytogenetic abnormalities. For illustrative purposes, the mechanism of action of lenalidomide can be subdivided into a cancer cell-intrinsic, a stromal, and an immunological component. Indeed, lenalidomide not only exerts direct cell cycle-arresting and pro-apoptotic effects on malignant cells, but also interferes with their physical and functional interaction with the tumor microenvironment and mediates a robust, pleiotropic immunostimulatory activity. In particular, lenalidomide has been shown to stimulate the cytotoxic functions of T lymphocytes and natural killer cells, to limit the immunosuppressive impact of regulatory T cells, and to modulate the secretion of a wide range of cytokines, including tumor necrosis factor α, interferon γ as well as interleukin (IL)-6, IL-10, and IL-12. Throughout the last decade, the antineoplastic and immunostimulatory potential of lenalidomide has been investigated in patients affected by a wide variety of hematological and solid malignancies. Here, we discuss the results of these studies and review the status of clinical trials currently assessing the safety and efficacy of this potent immunomodulatory drug in oncological indications.
Collapse
Affiliation(s)
- Michaela Semeraro
- Gustave Roussy; Villejuif, France ; INSERM, U1015, CICBT507; Villejuif, France
| | - Erika Vacchelli
- Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | - Jerome Galon
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U872; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015, CICBT507; Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France
| |
Collapse
|
38
|
Raychaudhuri N, Fernando R, Smith TJ. Thyrotropin regulates IL-6 expression in CD34+ fibrocytes: clear delineation of its cAMP-independent actions. PLoS One 2013; 8:e75100. [PMID: 24086448 PMCID: PMC3783445 DOI: 10.1371/journal.pone.0075100] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/11/2013] [Indexed: 11/18/2022] Open
Abstract
IL-6 plays diverse roles in normal and disease-associated immunity such as that associated with Graves’ disease (GD). In that syndrome, the orbit undergoes remodeling during a process known as thyroid-associated ophthalmopathy (TAO). Recently, CD34+ fibrocytes were found to infiltrate the orbit in TAO where they transition into CD34+ orbital fibroblasts. Surprisingly, fibrocytes display high levels of functional thyrotropin receptor (TSHR), the central antigen in GD. We report here that TSH and the pathogenic anti-TSHR antibodies that drive hyperthyroidism in GD induce IL-6 expression in fibrocytes and orbital fibroblasts. Unlike TSHR signaling in thyroid epithelium, that occurring in fibrocytes is completely independent of adenylate cyclase activation and cAMP generation. Instead TSH activates PDK1 and both AKT/PKB and PKC pathways. Expression and use of PKCβII switches to that of PKCµ as fibrocytes transition to TAO orbital fibroblasts. This shift is imposed by CD34− orbital fibroblasts but reverts when CD34+ fibroblasts are isolated. The up-regulation of IL-6 by TSH results from coordinately enhanced IL-6 gene promoter activity and increased IL-6 mRNA stability. TSH-dependent IL-6 expression requires activity at both CREB (−213 to −208 nt) and NF-κB (–78 to −62 nt) binding sites. These results provide novel insights into the molecular action of TSH and signaling downstream for TSHR in non-thyroid cells. Fibrocytes neither express adenylate cyclase nor generate cAMP and thus these findings are free from any influence of cAMP-related signaling. They identify potential therapeutic targets for TAO.
Collapse
Affiliation(s)
- Nupur Raychaudhuri
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Roshini Fernando
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Terry J. Smith
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
39
|
Ziakas PD, Karsaliakos P, Prodromou ML, Mylonakis E. Interleukin-6 polymorphisms and hematologic malignancy: a re-appraisal of evidence from genetic association studies. Biomarkers 2013; 18:625-31. [DOI: 10.3109/1354750x.2013.840799] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
Shain KH, Tao J. The B-cell receptor orchestrates environment-mediated lymphoma survival and drug resistance in B-cell malignancies. Oncogene 2013; 33:4107-13. [PMID: 24037527 DOI: 10.1038/onc.2013.379] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/24/2013] [Accepted: 07/29/2013] [Indexed: 12/14/2022]
Abstract
Specific niches within the lymphoma tumor microenvironment (TME) provide sanctuary for subpopulations of tumor cells through stromal cell-tumor cell interactions. These interactions notably dictate growth, response to therapy and resistance of residual malignant B cells to therapeutic agents. This minimal residual disease (MRD) remains a major challenge in the treatment of B-cell malignancies and contributes to subsequent disease relapse. B-cell receptor (BCR) signaling has emerged as essential mediator of B-cell homing, survival and environment-mediated drug resistance (EMDR). Central to EMDR are chemokine- and integrin-mediated interactions between lymphoma and the TME. Further, stromal cell-B cell adhesion confers a sustained BCR signaling leading to chemokine and integrin activation. Recently, the inhibitors of BCR signaling have garnered a substantial clinical interest because of their effectiveness in B-cell disorders. The efficacy of these agents is, at least in part, attributed to attenuation of BCR-dependent lymphoma-TME interactions. In this review, we discuss the pivotal role of BCR signaling in the integration of intrinsic and extrinsic determinants of TME-mediated lymphoma survival and drug resistance.
Collapse
Affiliation(s)
- K H Shain
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine Program, H Lee Moffitt Cancer Center and Research Institute at the University of South Florida, Tampa, FL, USA
| | - J Tao
- Departments of Hematopathology and Laboratory Medicine, and Chemical Biology and Molecular Medicine Program, H Lee Moffitt Cancer Center and Research Institute at the University of South Florida, Tampa, FL, USA
| |
Collapse
|
41
|
Mihou D, Katodritou E, Zervas K. Multiple myeloma staging based on the combination of beta-2-microglobulin and albumin: The role of albumin in the model. Hematology 2013; 12:527-31. [PMID: 17852450 DOI: 10.1080/10245330701384161] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Since the prognostic significance of the combination of beta-2-microglobulin (beta(2)m) and albumin in multiple myeloma (MM) has been recognized, these two easily obtainable parameters were subsequently employed in the staging systems of Bataille et al. (BSS), the South West Oncology Group (SWOG SS) and most recently the International Myeloma Working Group (ISS). There is no consensus, however, regarding the cut off levels of beta(2)m and the stage, early or advanced, at which albumin should be added to the model. At the same time, Weber et al. demonstrated similar results using beta(2)m alone in identical cut-offs with ISS (WSS). The aim of the present study is to apply these four staging systems in 504 MM patients, in order to discern the role of albumin in MM staging and evaluate if, and at which stage, albumin should be added to the model. METHODS Median overall survival (OS) according to BSS, SWOG SS, ISS and WSS was estimated according to the Kaplan-Meier method. OS differences between the stages were assessed using the log-rank test. Patients with beta(2)m < 3.5 mg/l and albumin < 3.5 g/dl, who were classified in stage II according to ISS and in stage I according to WSS, were analyzed separately in order to detect in which prognostic group they practically belong. RESULTS BSS and SWOG SS failed to distinguish stage II from stage III patients and stage III from stage IV patients, respectively. ISS and WSS achieved clear stratification of the patients into three distinct prognostic subgroups, but WSS I patients had a lower life expectancy than ISS I patients. This difference was due to false inclusion of patients with beta(2)m < 3.5 mg/l and albumin < 3.5 g/dl in stage I by WSS, while separate analysis of these patients proved that they belong, in fact, in stage II. In an attempt to improve its prognostic impact, WSS was then successfully modified by dividing WSS I patients in two substages, WSS IA: beta(2)m < 2.5 mg/l and WSS IB: 2.5 mg/l < or = beta(2)m < 3.5 mg/l, thus designating a low-risk and a low-intermediate-risk subgroup, respectively. CONCLUSION Albumin appears to lose its prognostic value at high cut-off levels of beta(2)m, while it enhances the prognostic significance of beta(2)m at low cut-off levels of the latter. Albumin cannot be eliminated from the ISS, since it is absolutely necessary in order to identify true low-risk patients. The only possibility for albumin exclusion from the model, could be to decrease the beta(2)m low-risk cut-off from 3.5 to 2.5 mg/l.
Collapse
Affiliation(s)
- Dimitra Mihou
- Department of Hematology-Oncology, Theagenion Cancer Center, Thessaloniki, Greece.
| | | | | |
Collapse
|
42
|
Corso A, Ferretti E, Lazzarino M. Zoledronic acid exerts its antitumor effect in multiple myeloma interfering with the bone marrow microenvironment. Hematology 2013; 10:215-24. [PMID: 16019470 DOI: 10.1080/10245330500094714] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by an excess of monotypic plasma cells which localize almost exclusively in the bone marrow provoking bone destruction via the activation of the osteoclasts. The bone marrow microenvironment, mainly through stromal cells, is strictly involved in the evolution of the disease supporting MM cell growth and survival [1]. MM plasma cells reside in the bone marrow by binding to adhesion molecule of extracellular matrix (ECM) and stromal cells. The activation of some signaling pathways within the stromal cells increases the production of several cytokines which in turn favors the myeloma cell proliferation and survival [2-6], and enhance the drug resistance by anti-apoptotic mechanisms [1,7-9]. Novel therapeutic agents target not only the myeloma cells but also the interaction between MM cells and the bone marrow microenvironment [8]. Bisphosphonates (Bps) interfere as well with bone microenvironment inhibiting the survival of stromal cells and hampering the contact between plasma and stromal cells. In this review we will revise preclinical evidences, and the potential mechanisms of the antitumor activity of zoledronic acid.
Collapse
Affiliation(s)
- Alessandro Corso
- Division of Hematology, IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.
| | | | | |
Collapse
|
43
|
Voorhees PM, Manges RF, Sonneveld P, Jagannath S, Somlo G, Krishnan A, Lentzsch S, Frank RC, Zweegman S, Wijermans PW, Orlowski RZ, Kranenburg B, Hall B, Casneuf T, Qin X, van de Velde H, Xie H, Thomas SK. A phase 2 multicentre study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with relapsed or refractory multiple myeloma. Br J Haematol 2013; 161:357-66. [PMID: 23432640 PMCID: PMC5837861 DOI: 10.1111/bjh.12266] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 01/17/2013] [Indexed: 11/30/2022]
Abstract
Interleukin-6 (IL6) plays a central role in multiple myeloma pathogenesis and confers resistance to corticosteroid-induced apoptosis. We therefore evaluated the efficacy and safety of siltuximab, an anti-IL6 monoclonal antibody, alone and in combination with dexamethasone, for patients with relapsed or refractory multiple myeloma who had ≥ 2 prior lines of therapy, one of which had to be bortezomib-based. Fourteen initial patients received siltuximab alone, 10 of whom had dexamethasone added for suboptimal response; 39 subsequent patients were treated with concurrent siltuximab and dexamethasone. Patients received a median of four prior lines of therapy, 83% were relapsed and refractory, and 70% refractory to their last dexamethasone-containing regimen. Suppression of serum C-reactive protein levels, a surrogate marker of IL6 inhibition, was demonstrated. There were no responses to siltuximab but combination therapy yielded a partial (17%) + minimal (6%) response rate of 23%, with responses seen in dexamethasone-refractory disease. The median time to progression, progression-free survival and overall survival for combination therapy was 4.4, 3.7 and 20.4 months respectively. Haematological toxicity was common but manageable. Infections occurred in 57% of combination-treated patients, including ≥ grade 3 infections in 18%. Further study of siltuximab in modern corticosteroid-containing myeloma regimens is warranted, with special attention to infection-related toxicity.
Collapse
Affiliation(s)
- Peter M Voorhees
- Division of Hematology-Oncology, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599-7305, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hernandez-Ilizaliturri FJ, Batoo SA. The emerging role of lenalidomide in the management of lymphoid malignancies. Ther Adv Hematol 2013; 2:45-53. [PMID: 23556075 DOI: 10.1177/2040620710390547] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Lenalidomide, a novel immunomodulatory drug (IMiD), is a promising therapeutic strategy for patients with relapsed/refractory chronic lymphocytic leukemia (CLL) and B-cell lymphomas. Biologically, the mechanisms responsible for lenalidomide activity are yet to be clearly defined. Based on preclinical models and early correlative studies conducted in parallel to clinical trials, lenalidomide has been found to enhance natural killer (NK)- and T-cell activity against tumor cells, alter the balance of pro- and anti-inflammatory cytokines in the tumor bed, inhibit angiogenesis, and, to a lesser degree, induce cell cycle arrest and apoptosis in cancer cells. Together, all of these biological effects appear to play a role in the activity observed in CLL or lymphoma patients treated with lenalidomide. Given the effect in NK- and T-cell function, lenalidomide is an alternative strategy to enhance the antitumor activity of monoclonal antibodies (mAbs). Clinical responses have been observed in patients with relapsed/refractory CLL, follicular lymphoma, small lymphocytic lymphoma, mantle cell lymphoma (MCL), and diffuse large B-cell lymphoma (DLBCL) treated with lenalidomide single agent. The favorable toxicity profile and route of administration made the use of lenalidomide an attractive therapy for certain types of patients (i.e. elderly, chemotherapy unfit, etc.). The erratic but serious incidence of tumor lysis syndrome and/or tumor flare reactions provides challenges in the incorporation of lenalidomide in the management of previously untreated CLL or CLL/lymphoma patients with bulky adenopathy. Correlative studies and/or retrospective analysis of lenalidomide-treated patients had identified several biomarkers associated with clinical endpoints in CLL (i.e. changes in tumor necrosis factor alpha [TNF-α] or vascular endothelial growth factor [VEGF] levels) or DLBCL (non-GCB phenotype) patients, but need to be validated. Early studies evaluating the efficacy and toxicity of lenalidomide in combination with rituximab in previously untreated indolent lymphoma are promising and warrant further study. In addition, the evaluation of lenalidomide in the maintenance setting or in combination with other target-specific agents (i.e. proteasome inhibitors) in aggressive lymphomas is being addressed in ongoing clinical trials. In summary, lenalidomide is emerging as a biologically active and novel agent in the treatment of B-cell neoplasms. Future translational and clinical studies will further define the role of lenalidomide in the management of de novo or relapsed/refractory CLL or B-cell lymphomas and identify the subset of patients most likely to gain clinical benefit.
Collapse
|
45
|
Molecular action of lenalidomide in lymphocytes and hematologic malignancies. Adv Hematol 2012; 2012:513702. [PMID: 22888354 PMCID: PMC3409527 DOI: 10.1155/2012/513702] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 05/12/2012] [Accepted: 06/18/2012] [Indexed: 01/10/2023] Open
Abstract
The immunomodulatory agent, lenalidomide, is a structural analogue of thalidomide approved by the US Food and Drug Administration for the treatment of myelodysplastic syndrome (MDS) and multiple myeloma (MM). This agent is also currently under active investigation for the treatment of chronic lymphocytic leukemia (CLL) and non-Hodgkin's lymphoma (NHL), as well as in drug combinations for some solid tumors and mantle cell lymphoma (MCL). Although treatment with lenalidomide has translated into a significant extension in overall survival in MM and MDS and has superior safety and efficacy relative to thalidomide, the mechanism of action as it relates to immune modulation remains elusive. Based on preclinical models and clinical trials, lenalidomide, as well as other structural thalidomide derivatives, enhances the proliferative and functional capacity of T-lymphocytes and amplifies costimulatory signaling pathways that activate effector responses and suppress inflammation. This paper summarizes our current understanding of T- and natural killer (NK) cell pathways that are modified by lenalidomide in hematopoietic neoplasms to inform future decisions about potential combination therapies.
Collapse
|
46
|
Lee SW, Cho HY, Na G, Yoo MR, Seo SK, Hur DY, Han J, Lee CK, Choi I. CD40 stimulation induces vincristine resistance via AKT activation and MRP1 expression in a human multiple myeloma cell line. Immunol Lett 2012; 144:41-8. [PMID: 22445357 DOI: 10.1016/j.imlet.2012.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/15/2012] [Accepted: 03/04/2012] [Indexed: 12/26/2022]
Abstract
Various co-stimulatory receptors are expressed in multiple myeloma (MM) both in immune microenvironment and in the tumor microenvironment in vivo. In relapsed human MM, these receptors are known to increase cell proliferation and induce conventional drug resistance. However, the mechanism of drug resistance induced via co-stimulatory receptors is poorly understood. In this study, we examined the role of CD40 expressed on MM cell lines. Out of all of the KMS MM cell lines, the KMS28BM cells expressed high levels of the CD40 receptor. When stimulated with anti-CD40 antibody or recombinant human CD40L, the proliferation of KMS28BM cells was increased 1.7 fold. In CD40-stimulated KMS28BM cells, signaling via the AKT pathway caused an increase in the expression of multidrug resistance-associated gene 1 (MRP1) and IL-6 by 2.2 fold and 30 fold, respectively, but not the MDR1 gene. Furthermore, CD40-stimulated KMS28BM cells were observed to be substantially resistant to the anticancer drug vincristine, and when cells were treated with the MRP1 specific inhibitor, MK-571, drug resistance was decreased. We also found that CD40-stimulated, MRP1-expressing KMS28BM cells significantly increased calcein efflux, and calcein efflux was inhibited through treatment with MK-571. Therefore, blocking CD40 and inhibiting MRP1 are potential targets to treat CD40-induced drug resistance in multiple myeloma.
Collapse
Affiliation(s)
- Soo-Woong Lee
- Advanced Research Center for Multiple Myeloma, College of Medicine, Inje University, Busan, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ciarcia R, Vitiello MT, Galdiero M, Pacilio C, Iovane V, d'Angelo D, Pagnini D, Caparrotti G, Conti D, Tomei V, Florio S, Giordano A. Imatinib treatment inhibit IL-6, IL-8, NF-KB and AP-1 production and modulate intracellular calcium in CML patients. J Cell Physiol 2012; 227:2798-803. [DOI: 10.1002/jcp.23029] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Bladé J, de Larrea CF, Rosiñol L. Incorporating monoclonal antibodies into the therapy of multiple myeloma. J Clin Oncol 2012; 30:1904-6. [PMID: 22291081 DOI: 10.1200/jco.2011.40.4178] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
49
|
Nair JR, Carlson LM, Koorella C, Rozanski CH, Byrne GE, Bergsagel PL, Shaughnessy JP, Boise LH, Chanan-Khan A, Lee KP. CD28 expressed on malignant plasma cells induces a prosurvival and immunosuppressive microenvironment. THE JOURNAL OF IMMUNOLOGY 2011; 187:1243-53. [PMID: 21715687 DOI: 10.4049/jimmunol.1100016] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Interactions between the malignant plasma cells of multiple myeloma and stromal cells within the bone marrow microenvironment are essential for myeloma cell survival, mirroring the same dependence of normal bone marrow-resident long-lived plasma cells on specific marrow niches. These interactions directly transduce prosurvival signals to the myeloma cells and also induce niche production of supportive soluble factors. However, despite their central importance, the specific molecular and cellular components involved remain poorly characterized. We now report that the prototypic T cell costimulatory receptor CD28 is overexpressed on myeloma cells during disease progression and in the poor-prognosis subgroups and plays a previously unrecognized role as a two-way molecular bridge to support myeloid stromal cells in the microenvironment. Engagement by CD28 to its ligand CD80/CD86 on stromal dendritic cell directly transduces a prosurvival signal to myeloma cell, protecting it against chemotherapy and growth factor withdrawal-induced death. Simultaneously, CD28-mediated ligation of CD80/CD86 induces the stromal dendritic cell to produce the prosurvival cytokine IL-6 (involving novel cross-talk with the Notch pathway) and the immunosuppressive enzyme IDO. These findings identify CD28 and CD80/CD86 as important molecular components of the interaction between myeloma cells and the bone marrow microenvironment, point to similar interaction for normal plasma cells, and suggest novel therapeutic strategies to target malignant and pathogenic (e.g., in allergy and autoimmunity) plasma cells.
Collapse
Affiliation(s)
- Jayakumar R Nair
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14226, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Saloura V, Grivas PD. Lenalidomide: a synthetic compound with an evolving role in cancer management. ACTA ACUST UNITED AC 2011; 15:318-31. [PMID: 20863427 DOI: 10.1179/102453310x12647083620921] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Lenalidomide is a functional and structural analogue of thalidomide with a relatively benign toxicity profile and pleiotropic anti-tumor activity, exhibiting anti-angiogenic and immunomodulatory effects. Lenalidomide has already been approved for the management of low or intermediate-1 risk myelodysplastic syndrome with chromosome 5q31 deletion and relapsed/refractory multiple myeloma in combination with dexamethasone. During the last five years, multiple clinical trials have been conducted to explore its potential efficacy in hematologic as well as in solid malignancies, revealing a significant benefit in clinical outcomes. This review outlines the mechanisms of action, the toxicity profile and the efficacy of lenalidomide, reviewing the current literature and focusing on the current status of the compound in cancer management. The determination of molecular biomarkers, predictive of clinical response to lenalidomide may reveal a more substantial role of this agent in the treatment of hematologic as well as solid malignancies.
Collapse
Affiliation(s)
- Vassiliki Saloura
- Department of Internal Medicine, Drexel University College of Medicine/Hahnemann University Hospital, Philadelphia, PA, USA
| | | |
Collapse
|