1
|
Lohse I, Dutcher G, Al-Ali H, Alperstein W, Coulter DW, Trucco M, Trent JC, Wahlestedt C. Drug Sensitivity Testing in Osteosarcoma: A Case Report. Curr Oncol 2025; 32:271. [PMID: 40422530 PMCID: PMC12110314 DOI: 10.3390/curroncol32050271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/14/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
Precision medicine approaches using ex-vivo drug sensitivity testing (DST) have received attention in the cancer research community as a means to improve treatment stratification in populations where multiple treatment attempts are not feasible, or no standard-of-care treatment exists, such as ultra-rare cancers with a significant clinical need for effective treatment options, like osteosarcoma. DST has the potential to supplement existing patient stratification approaches by providing tumor-specific response data to aid in treatment selection at the time of treatment decision. We present the case of a pediatric osteosarcoma patient who was evaluated using DST at the time of standard-of-care treatment to evaluate treatment sensitivity. The DST screen indicated significant treatment sensitivity to anthracyclines and methotrexate, consistent with the first-line standard-of-care therapy (MAP). Clinical follow-up showed treatment sensitivity to standard-of-care MAP treatment and pathology results of 90% necrosis. The present case shows that DST screening is feasible from a technical standpoint, can be performed in a clinically relevant time frame that does not delay treatment start, and provides personalized drug sensitivity information on clinically available agents, and the DST results align with the clinical treatment response.
Collapse
Affiliation(s)
- Ines Lohse
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Molecular Therapeutics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Giselle Dutcher
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (G.D.); (H.A.-A.); (W.A.); (M.T.); (J.C.T.)
| | - Hassan Al-Ali
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (G.D.); (H.A.-A.); (W.A.); (M.T.); (J.C.T.)
- Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Warren Alperstein
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (G.D.); (H.A.-A.); (W.A.); (M.T.); (J.C.T.)
| | - Donald W. Coulter
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Matteo Trucco
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (G.D.); (H.A.-A.); (W.A.); (M.T.); (J.C.T.)
| | - Jonathan C. Trent
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (G.D.); (H.A.-A.); (W.A.); (M.T.); (J.C.T.)
| | - Claes Wahlestedt
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
2
|
Li L, Hashemi L, Eid J, Tao W, Campoverde L, Yu A, Farooqi AA, Al-Ali H, D'Amato G, Hornicek F, Duan Z, Lohse I, Trent J. High-Throughput Drug Screening in Chondrosarcoma Cells Identifies Effective Antineoplastic Agents Independent of IDH Mutation. Int J Mol Sci 2024; 25:13003. [PMID: 39684713 DOI: 10.3390/ijms252313003] [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: 10/14/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
The term chondrosarcoma refers to a rare and heterogeneous group of malignant cartilaginous tumors that are typically resistant to chemotherapy and radiotherapy. Metastatic chondrosarcoma has a poor prognosis, and effective systemic therapies are lacking. Isocitrate dehydrogenase (IDH) mutations represent a potential therapeutic target, but IDH inhibitors alone have shown limited clinical efficacy to date. Although the role of conventional chemotherapy is still subject to debate, some evidence suggests it may provide therapeutic benefits in advanced cases. In this study, we aimed to identify effective compounds for combination therapy in chondrosarcoma. Using high-throughput screening, we evaluated a panel of anticancer agents in IDH1-mutant chondrosarcoma cell lines and their mutant IDH1 knockout derivatives. The top 20 most potent compounds were identified across all cell lines, irrespective of IDH mutation status. Representative drugs selected for further investigation included docetaxel, methotrexate, panobinostat, idarubicin, camptothecin, and pevonedistat. These drugs inhibited colony formation, induced apoptosis and cell cycle arrest, and exhibited synergistic antitumor activity in two-drug combinations. In conclusion, we identified several highly effective agents with potent anti-tumor activity in chondrosarcoma cells, independent of IDH mutation status. These agents represent promising candidates for chondrosarcoma therapy and warrant further preclinical investigation and potential inclusion in clinical trials.
Collapse
Affiliation(s)
- Luyuan Li
- Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
| | - Lily Hashemi
- College of Science, Northeastern University, Boston, MA 02115, USA
| | - Josiane Eid
- Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
| | - Wensi Tao
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Leticia Campoverde
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amy Yu
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | - Hassan Al-Ali
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Frost Institute for Data Science and Computing, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Gina D'Amato
- Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
| | - Francis Hornicek
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
- Department of Orthopedics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zhenfeng Duan
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
- Department of Orthopedics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ines Lohse
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jonathan Trent
- Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, USA
| |
Collapse
|
3
|
祁 文, 徐 学, 李 白, 王 叨, 盛 光, 朱 平, 王 春. [Application of high-throughput drug sensitivity testing in children with relapsed and refractory acute leukemia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:1093-1100. [PMID: 39467680 PMCID: PMC11527403 DOI: 10.7499/j.issn.1008-8830.2406014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/09/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVES To explore the current application of high-throughput drug sensitivity (HDS) testing in children with relapsed and refractory acute leukemia (RR-AL) and analyze the feasibility of salvage treatment plans. METHODS A retrospective collection of clinical data from children with RR-AL who underwent HDS testing at the Department of Children's Hematology and Oncology of the First Affiliated Hospital of Zhengzhou University from November 2021 to October 2023 was conducted, followed by an analysis of drug sensitivity results and treatment outcomes. RESULTS A total of 17 children with RR-AL underwent HDS testing, including 7 cases of relapsed refractory acute myeloid leukemia and 10 cases of relapsed refractory acute lymphoblastic leukemia. The detection rate of highly sensitive chemotherapy drugs/regimens was 53% (9/17), while the detection rate of moderately sensitive chemotherapy drugs/regimens was 100% (17/17). Among the 17 RR-AL patients with highly and moderately sensitive chemotherapy drugs and regimens, the MOACD regimen (mitoxantrone + vincristine + cytarabine + cyclophosphamide + dexamethasone) accounted for 100%, with the highest inhibition rate for single-agent mitoxantrone (94%, 16/17), and the highest inhibition rate for targeted therapy being bortezomib (94%, 16/17). Nine patients adjusted their chemotherapy based on HDS testing results, with 4 undergoing hematopoietic stem cell transplantation. Four patients achieved disease-free survival, while 5 died. Eight patients received empirical chemotherapy, with 2 undergoing hematopoietic stem cell transplantation; 4 achieved disease-free survival, while 4 died. CONCLUSIONS HDS testing can identify highly sensitive drugs/regimens for children with RR-AL, improving the rate of re-remission and creating conditions for subsequent hematopoietic stem cell transplantation.
Collapse
|
4
|
Garbuzenko OB, Sapiezynski J, Girda E, Rodriguez-Rodriguez L, Minko T. Personalized Versus Precision Nanomedicine for Treatment of Ovarian Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307462. [PMID: 38342698 PMCID: PMC11316847 DOI: 10.1002/smll.202307462] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/22/2023] [Indexed: 02/13/2024]
Abstract
The response to treatment is substantially varied between individual patients with ovarian cancer. However, chemotherapy treatment plans rarely pay sufficient attention to the mentioned factors. Instead, standardized treatment protocols are usually employed for most ovarian cancer patients. Variations in an individual's sensitivity to drugs significantly limit the effectiveness of treatment in some patients and lead to severe toxicities in others. In the present investigation, a nanotechnology-based approach for personalized treatment of ovarian carcinoma (the most lethal type of gynecological cancer) constructed on the individual genetic profile of the patient's tumor is developed and validated. The expression of predefined genes and proteins is analyzed for each patient sample. Finally, a mixture of the complex nanocarrier-based targeted delivery system containing drug(s)/siRNA(s)/targeted peptide is selected from the pre-synthesized bank and tested in vivo on murine cancer model using cancer cells isolated from tumors of each patient. Based on the results of the present study, an innovative approach and protocol for personalized treatment of ovarian cancer are suggested and evaluated. The results of the present study clearly show the advantages and perspectives of the proposed individual treatment approach.
Collapse
Affiliation(s)
- Olga B. Garbuzenko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, Piscataway, NJ USA 08854
| | - Justin Sapiezynski
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, Piscataway, NJ USA 08854
| | - Eugenia Girda
- Department of Gynecology Oncology, Robert Wood Johnson School of Medicine, Rutgers the State University of New Jersey, New Brunswick, NJ USA 08901
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA 08901
| | - Lorna Rodriguez-Rodriguez
- Department of Surgery, Division of Gynecologic Oncology, City of Hope National Medical Center, Duarte, CA 91010
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, Piscataway, NJ USA 08854
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA 08901
| |
Collapse
|
5
|
Therapeutic strategies for non-small cell lung cancer: Experimental models and emerging biomarkers to monitor drug efficacies. Pharmacol Ther 2023; 242:108347. [PMID: 36642389 DOI: 10.1016/j.pharmthera.2023.108347] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
While new targeted therapies have considerably changed the treatment and prognosis of non-small cell lung cancer (NSCLC), they are frequently unsuccessful due to primary or acquired resistances. Chemoresistance is a complex process that combines cancer cell intrinsic mechanisms including molecular and genetic abnormalities, aberrant interactions within the tumor microenvironment, and the pharmacokinetic characteristics of each molecule. From a pharmacological point of view, two levers could improve the response to treatment: (i) developing tools to predict the response to chemo- and targeted therapies and (ii) gaining a better understanding of the influence of the tumor microenvironment. Both personalized medicine approaches require the identification of relevant experimental models and biomarkers to understand and fight against chemoresistance mechanisms. After describing the main therapies in NSCLC, the scope of this review will be to identify and to discuss relevant in vitro and ex vivo experimental models that are able to mimic tumors. In addition, the interests of these models in the predictive responses to proposed therapies will be discussed. Finally, this review will evaluate the involvement of novel secreted biomarkers such as tumor DNA or micro RNA in predicting responses to anti-tumor therapies.
Collapse
|
6
|
Szklener K, Michalski A, Żak K, Piwoński M, Mańdziuk S. Ibrutinib in the Treatment of Solid Tumors: Current State of Knowledge and Future Directions. Cells 2022; 11:1338. [PMID: 35456016 PMCID: PMC9032968 DOI: 10.3390/cells11081338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Bruton's Tyrosine Kinase (BTK) is considered crucial in the activation and survival of both physiological and malignant B-cells. In recent years, ibrutinib, an oral BTK inhibitor, became a breakthrough therapy for hematological malignancies, such as chronic lymphocytic. However, ibrutinib's feasibility might not end there. Several other kinases with established involvement with solid malignancies (i.e., EGFR, HER2) have been found to be inhibited by this agent. Recent discoveries indicate that BTK is a potential anti-solid tumor therapy target. Consequently, ibrutinib, a BTK-inhibitor, has been studied as a therapeutic option in solid malignancies. While most preclinical studies indicate ibrutinib to be an effective therapeutic option in some specific indications, such as NSCLC and breast cancer, clinical trials contradict these observations. Nevertheless, while ibrutinib failed as a monotherapy, it might become an interesting part of a multidrug regime: not only has a synergism between ibrutinib and other compounds, such as trametinib or dactolisib, been observed in vitro, but this BTK inhibitor has also been established as a radio- and chemosensitizer. This review aims to describe the milestones in translating BTK inhibitors to solid tumors in order to understand the future potential of this agent better.
Collapse
Affiliation(s)
- Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 20-090 Lublin, Poland; (A.M.); (K.Ż.); (M.P.); (S.M.)
| | | | | | | | | |
Collapse
|
7
|
Acanda De La Rocha AM, Fader M, Coats ER, Espinal PS, Berrios V, Saghira C, Sotto I, Shakya R, Janvier M, Khatib Z, Abdella H, Bittle M, Andrade-Feraud CM, Guilarte TR, McCafferty-Fernandez J, Salyakina D, Azzam DJ. Clinical Utility of Functional Precision Medicine in the Management of Recurrent/Relapsed Childhood Rhabdomyosarcoma. JCO Precis Oncol 2021; 5:PO.20.00438. [PMID: 34738048 PMCID: PMC8563073 DOI: 10.1200/po.20.00438] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 08/12/2021] [Accepted: 09/24/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Arlet M Acanda De La Rocha
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL
| | - Maggie Fader
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL.,Pediatric Oncology and Hematology, Nicklaus Children's Hospital, Miami, FL
| | - Ebony R Coats
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL
| | - Paula S Espinal
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL
| | - Vanessa Berrios
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL
| | - Cima Saghira
- Miller School of Medicine, University of Miami, Miami, FL
| | - Ileana Sotto
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL
| | - Rojesh Shakya
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL
| | - Michelin Janvier
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL
| | - Ziad Khatib
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL.,Pediatric Oncology and Hematology, Nicklaus Children's Hospital, Miami, FL
| | - Haneen Abdella
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL.,Pediatric Oncology and Hematology, Nicklaus Children's Hospital, Miami, FL
| | - Mathew Bittle
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL
| | - Cristina M Andrade-Feraud
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL
| | | | - Daria Salyakina
- Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL
| | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL
| |
Collapse
|
8
|
Khalafi S, Zhu S, Khurana R, Lohse I, Giordano S, Corso S, Al-Ali H, Brothers SP, Wahlestedt C, Schürer S, El-Rifai W. A novel strategy for combination of clofarabine and pictilisib is synergistic in gastric cancer. Transl Oncol 2021; 15:101260. [PMID: 34735897 PMCID: PMC8571525 DOI: 10.1016/j.tranon.2021.101260] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/08/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Drug sensitivity testing identified novel drugs like clofarabine effective in treating gastric cancer. mRNA sequencing can be used to identify agents with synergistic activity to a reference compound. Pictilisib sensitizes gastric cancer to clofarabine treatment through AKT inhibition. The combination of clofarabine and pictilisib inhibits tumor growth in cell lines and PDX models.
Gastric cancer (GC) is frequently characterized by resistance to standard chemotherapeutic regimens and poor clinical outcomes. We aimed to identify a novel therapeutic approach using drug sensitivity testing (DST) and our computational SynerySeq pipeline. DST of GC cell lines was performed with a library of 215 Federal Drug Administration (FDA) approved compounds and identified clofarabine as a potential therapeutic agent. RNA-sequencing (RNAseq) of clofarabine treated GC cells was analyzed according to our SynergySeq pipeline and identified pictilisib as a potential synergistic agent. Clonogenic survival and Annexin V assays demonstrated increased cell death with clofarabine and pictilisib combination treatment (P<0.01). The combination induced double strand breaks (DSB) as indicated by phosphorylated H2A histone family member X (γH2AX) immunofluorescence and western blot analysis (P<0.01). Pictilisib treatment inhibited the protein kinase B (AKT) cell survival pathway and promoted a pro-apoptotic phenotype as evidenced by quantitative real time polymerase chain reaction (qRT-PCR) analysis of the B-cell lymphoma 2 (BCL2) protein family members (P<0.01). Patient derived xenograft (PDX) data confirmed that the combination is more effective in abrogating tumor growth with prolonged survival than single-agent treatment (P<0.01). The novel combination of clofarabine and pictilisib in GC promotes DNA damage and inhibits key cell survival pathways to induce cell death beyond single-agent treatment.
Collapse
Affiliation(s)
- Shayan Khalafi
- Department of Surgery, Miller School of Medicine, University of Miami, Rosenstiel Medical Science Bldg, 1600 NW 10th Ave, Room 4007, Miami, FL 33136-1015, United States
| | - Shoumin Zhu
- Department of Surgery, Miller School of Medicine, University of Miami, Rosenstiel Medical Science Bldg, 1600 NW 10th Ave, Room 4007, Miami, FL 33136-1015, United States; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Rimpi Khurana
- Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Ines Lohse
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Molecular Therapeutics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami, FL 33136, United States
| | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo 10060, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo 10060, Italy
| | - Simona Corso
- Department of Oncology, University of Torino, Candiolo 10060, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo 10060, Italy
| | - Hassan Al-Ali
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Peggy and Harold Katz Drug Discovery Center, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Shaun P Brothers
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Claes Wahlestedt
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Stephan Schürer
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Institute for Data Science and Computing, University of Miami, Miami, FL 33136, United States
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Rosenstiel Medical Science Bldg, 1600 NW 10th Ave, Room 4007, Miami, FL 33136-1015, United States; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Veterans Affairs, Miami Healthcare System, Miami, FL 33136, United States.
| |
Collapse
|
9
|
Metzler JM, Burla L, Fink D, Imesch P. Ibrutinib in Gynecological Malignancies and Breast Cancer: A Systematic Review. Int J Mol Sci 2020; 21:ijms21114154. [PMID: 32532074 PMCID: PMC7312555 DOI: 10.3390/ijms21114154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/02/2020] [Accepted: 06/07/2020] [Indexed: 12/14/2022] Open
Abstract
Ibrutinib is an orally available, small-molecule tyrosine kinase inhibitor. Its main purpose is to inhibit Bruton's tyrosine kinase (BTK), an enzyme that is crucial in B cell development. It is FDA approved for the treatment of certain hematological malignancies. Several promising off-target drug effects have led to multiple, mostly preclinical investigations regarding its use in solid tumors. Unfortunately, data on its effectiveness in gynecological malignancies are limited, and (systematic) reviews are missing. The objective of this review was to summarize the existing literature and to analyze the evidence of ibrutinib as a treatment option in gynecological malignancies, including breast cancer. Studies were identified in MEDLINE and EMBASE using a defined search strategy, and preclinical or clinical research projects investigating ibrutinib in connection with these malignancies were considered eligible for inclusion. Our findings showed that preclinical studies generally confirm ibrutinib's efficacy in cell lines and animal models of ovarian, breast, and endometrial cancer. Ibrutinib exerts multiple antineoplastic effects, such as on-target BTK inhibition, off-target kinase inhibition, and immunomodulation by interference with myeloid-derived suppressor cells (MDSCs), programmed death-ligand 1 (PD-L1), and T cell response. These mechanisms were elaborated and discussed in the context of the evidence available. Further research is needed in order to transfer the preclinical results to a broader clinical appliance.
Collapse
|
10
|
Qiu L, Wang J, Chen M, Chen F, Tu W. Exosomal microRNA‑146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2‑mediated PI3K/Akt axis. Int J Mol Med 2020; 46:609-620. [PMID: 32626953 PMCID: PMC7307828 DOI: 10.3892/ijmm.2020.4634] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
The carrier role of exosomes from human umbilical cord mesenchymal stem cells (hUCMSCs) containing microRNAs (miRNAs) has been implicated in gene and drug therapy. The aim of the present study was to investigate the role of exosomal microRNA-146a (miR-146a) from hUCMSCs in ovarian cancer (OC). Following the generation of docetaxel (DTX)-resistant SKOV3 cells and taxane-resistant A2780 cells, exosomes were isolated from hUCMSCs and added to the chemoresistant cells. Microarray analysis revealed that miR-146a expression was upregulated in DTX/SKOV3 cells among 15 ectopically expressed miRNAs. Analysis using the StarBase and miRSearch databases demonstrated that miR-146a targeted laminin γ2 (LAMC2), which was further verified using dual-luciferase reporter assays. Subsequently, miR-146a inhibitor or LAMC2 overexpression vectors were transfected into hUCMSCs or OC cells, respectively, and their effects on growth and chemoresistance in OC cells were assessed. The hUCMSC-derived exosomes reduced cell growth and chemoresistance in OC. Furthermore, hUCMSC-derived exosomes with miR-146a expression knocked down increased OC cell growth and chemoresistance, which was mediated by the PI3K/Akt signaling pathway via LAMC2.
Collapse
Affiliation(s)
- Liya Qiu
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Jiakun Wang
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Mei Chen
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Fengyun Chen
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Wenluo Tu
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital, Taizhou, Zhejiang 318000, P.R. China
| |
Collapse
|