24 results on '"Chen Hao Lo"'
Search Results
2. Computational modeling reveals a key role for polarized myeloid cells in controlling osteoclast activity during bone injury repair
- Author
-
Chen Hao Lo, Etienne Baratchart, David Basanta, and Conor C. Lynch
- Subjects
Medicine ,Science - Abstract
Abstract Bone-forming osteoblasts and -resorbing osteoclasts control bone injury repair, and myeloid-derived cells such as monocytes and macrophages are known to influence their behavior. However, precisely how these multiple cell types coordinate and regulate each other over time within the bone marrow to restore bone is difficult to dissect using biological approaches. Conversely, mathematical modeling lends itself well to this challenge. Therefore, we generated an ordinary differential equation (ODE) model powered by experimental data (osteoblast, osteoclast, bone volume, pro- and anti-inflammatory myeloid cells) obtained from intra-tibially injured mice. Initial ODE results using only osteoblast/osteoclast populations demonstrated that bone homeostasis could not be recovered after injury, but this issue was resolved upon integration of pro- and anti-inflammatory myeloid population dynamics. Surprisingly, the ODE revealed temporal disconnects between the peak of total bone mineralization/resorption, and osteoblast/osteoclast numbers. Specifically, the model indicated that osteoclast activity must vary greatly (> 17-fold) to return the bone volume to baseline after injury and suggest that osteoblast/osteoclast number alone is insufficient to predict bone the trajectory of bone repair. Importantly, the values of osteoclast activity fall within those published previously. These data underscore the value of mathematical modeling approaches to understand and reveal new insights into complex biological processes.
- Published
- 2021
- Full Text
- View/download PDF
3. Mesenchymal stem cell-derived interleukin-28 drives the selection of apoptosis resistant bone metastatic prostate cancer
- Author
-
Jeremy J. McGuire, Jeremy S. Frieling, Chen Hao Lo, Tao Li, Ayaz Muhammad, Harshani R. Lawrence, Nicholas J. Lawrence, Leah M. Cook, and Conor C. Lynch
- Subjects
Science - Abstract
The effects of bone-marrow derived MSCs on prostate cancer cells remain unknown. Here the authors show that MSC-derived IL-28 induces prostate cancer cell apoptosis via IL-28Rα-STAT1 signalling, while acquired resistance to apoptosis is associated with a shift in IL-28Rα signalling via STAT1 to STAT3.
- Published
- 2021
- Full Text
- View/download PDF
4. Integrated computational and in vivo models reveal Key Insights into macrophage behavior during bone healing.
- Author
-
Etienne Baratchart, Chen Hao Lo, Conor C Lynch, and David Basanta
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Myeloid-derived monocyte and macrophages are key cells in the bone that contribute to remodeling and injury repair. However, their temporal polarization status and control of bone-resorbing osteoclasts and bone-forming osteoblasts responses is largely unknown. In this study, we focused on two aspects of monocyte/macrophage dynamics and polarization states over time: 1) the injury-triggered pro- and anti-inflammatory monocytes/macrophages temporal profiles, 2) the contributions of pro- versus anti-inflammatory monocytes/macrophages in coordinating healing response. Bone healing is a complex multicellular dynamic process. While traditional in vitro and in vivo experimentation may capture the behavior of select populations with high resolution, they cannot simultaneously track the behavior of multiple populations. To address this, we have used an integrated coupled ordinary differential equations (ODEs)-based framework describing multiple cellular species to in vivo bone injury data in order to identify and test various hypotheses regarding bone cell populations dynamics. Our approach allowed us to infer several biological insights including, but not limited to,: 1) anti-inflammatory macrophages are key for early osteoclast inhibition and pro-inflammatory macrophage suppression, 2) pro-inflammatory macrophages are involved in osteoclast bone resorptive activity, whereas osteoblasts promote osteoclast differentiation, 3) Pro-inflammatory monocytes/macrophages rise during two expansion waves, which can be explained by the anti-inflammatory macrophages-mediated inhibition phase between the two waves. In addition, we further tested the robustness of the mathematical model by comparing simulation results to an independent experimental dataset. Taken together, this novel comprehensive mathematical framework allowed us to identify biological mechanisms that best recapitulate bone injury data and that explain the coupled cellular population dynamics involved in the process. Furthermore, our hypothesis testing methodology could be used in other contexts to decipher mechanisms in complex multicellular processes.
- Published
- 2022
- Full Text
- View/download PDF
5. Multifaceted Roles for Macrophages in Prostate Cancer Skeletal Metastasis
- Author
-
Chen Hao Lo and Conor C. Lynch
- Subjects
bone ,prostate cancer ,metastasis ,macrophage ,polarization ,therapy ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Bone-metastatic prostate cancer is common in men with recurrent castrate-resistant disease. To date, therapeutic focus has largely revolved around androgen deprivation therapy (ADT) and chemotherapy. While second-generation ADTs and combination ADT/chemotherapy approaches have been successful in extending overall survival, the disease remains incurable. It is clear that molecular and cellular components of the cancer-bone microenvironment contribute to the disease progression and potentially to the emergence of therapy resistance. In bone, metastatic prostate cancer cells manipulate bone-forming osteoblasts and bone-resorbing osteoclasts to produce growth and survival factors. While osteoclast-targeted therapies such as bisphosphonates have improved quality of life, emerging data have defined important roles for additional cells of the bone microenvironment, including macrophages and T cells. Disappointingly, early clinical trials with checkpoint blockade inhibitors geared at promoting cytotoxic T cell response have not proved as promising for prostate cancer compared to other solid malignancies. Macrophages, including bone-resident osteomacs, are a major component of the bone marrow and play key roles in coordinating normal bone remodeling and injury repair. The role for anti-inflammatory macrophages in the progression of primary prostate cancer is well established yet relatively little is known about macrophages in the context of bone-metastatic prostate cancer. The focus of the current review is to summarize our knowledge of macrophage contribution to normal bone remodeling and prostate-to-bone metastasis, while also considering the impact of standard of care and targeted therapies on macrophage behavior in the tumor-bone microenvironment.
- Published
- 2018
- Full Text
- View/download PDF
6. γδ-Enriched CAR-T cell therapy for bone metastatic castrate-resistant prostate cancer
- Author
-
Jeremy S. Frieling, Leticia Tordesillas, Xiomar E. Bustos, Maria Cecilia Ramello, Ryan T. Bishop, Junior E. Cianne, Sebastian A. Snedal, Tao Li, Chen Hao Lo, Janis de la Iglesia, Emiliano Roselli, Ismahène Benzaïd, Xuefeng Wang, Youngchul Kim, Conor C. Lynch, and Daniel Abate-Daga
- Subjects
Multidisciplinary - Abstract
Immune checkpoint blockade has been largely unsuccessful for the treatment of bone metastatic castrate-resistant prostate cancer (mCRPC). Here, we report a combinatorial strategy to treat mCRPC using γδ-enriched chimeric antigen receptor (CAR) T cells and zoledronate (ZOL). In a preclinical murine model of bone mCRPC, γδ CAR-T cells targeting prostate stem cell antigen (PSCA) induced a rapid and significant regression of established tumors, combined with increased survival and reduced cancer-associated bone disease. Pretreatment with ZOL, a U.S. Food and Drug Administration–approved bisphosphonate prescribed to mitigate pathological fracture in mCRPC patients, resulted in CAR-independent activation of γδ CAR-T cells, increased cytokine secretion, and enhanced antitumor efficacy. These data show that the activity of the endogenous Vγ9Vδ2 T cell receptor is preserved in CAR-T cells, allowing for dual-receptor recognition of tumor cells. Collectively, our findings support the use of γδ CAR-T cell therapy for mCRPC treatment.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Data from Autocrine TGFβ Is a Survival Factor for Monocytes and Drives Immunosuppressive Lineage Commitment
- Author
-
Mary Helen Barcellos-Hoff, Matthew H. Spitzer, Diana M. Marquez, Iliana Tenvooren, Renate Parry, Ritu Roy, Chen Hao Lo, Shisuo Du, Ilenia Pellicciotta, Kyla E. Driscoll, and Alba Gonzalez-Junca
- Abstract
Supplemental tables and figures
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Autocrine TGFβ Is a Survival Factor for Monocytes and Drives Immunosuppressive Lineage Commitment
- Author
-
Mary Helen Barcellos-Hoff, Matthew H. Spitzer, Diana M. Marquez, Iliana Tenvooren, Renate Parry, Ritu Roy, Chen Hao Lo, Shisuo Du, Ilenia Pellicciotta, Kyla E. Driscoll, and Alba Gonzalez-Junca
- Abstract
Transforming growth factor β (TGFβ) is an effector of immune suppression and contributes to a permissive tumor microenvironment that compromises effective immunotherapy. We identified a correlation between TGFB1 and genes expressed by myeloid cells, but not granulocytes, in The Cancer Genome Atlas lung adenocarcinoma data, in which high TGFB1 expression was associated with poor survival. To determine whether TGFβ affected cell fate decisions and lineage commitment, we studied primary cultures of CD14+ monocytes isolated from peripheral blood of healthy donors. We discovered that TGFβ was a survival factor for CD14+ monocytes, which rapidly executed an apoptotic program in its absence. Continued exposure to TGFβ in combination with granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin 6 (IL6) amplified HLA-DRlowCD14+CD11b+CD33+ myeloid-derived suppressor cells (MDSCs) at the expense of macrophage and dendritic cell (DC) differentiation. MDSCs generated in the presence of TGFβ were more effective in suppressing T-cell proliferation and promoted the T regulatory cell phenotype. In contrast, inhibition of TGFβ signaling using a small-molecule inhibitor of receptor kinase activity in CD14+ monocytes treated with GM-CSF and IL6 decreased MDSC differentiation and increased differentiation to proinflammatory macrophages and antigen-presenting DCs. The effect of autocrine and paracrine TGFβ on myeloid cell survival and lineage commitment suggests that pharmacologic inhibition of TGFβ-dependent signaling in cancer would favor antitumor immunity.
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Host-Derived Matrix Metalloproteinase-13 Activity Promotes Multiple Myeloma–Induced Osteolysis and Reduces Overall Survival
- Author
-
Conor C. Lynch, Gregg B. Fields, Anna M. Knapinska, William R. Roush, Rita Fuerst, Jun Yong Choi, Kenneth H. Shain, Tao Li, Jeremy J. McGuire, Gemma Shay, and Chen Hao Lo
- Abstract
Multiple myeloma promotes systemic skeletal bone disease that greatly contributes to patient morbidity. Resorption of type I collagen–rich bone matrix by activated osteoclasts results in the release of sequestered growth factors that can drive progression of the disease. Matrix metalloproteinase-13 (MMP13) is a collagenase expressed predominantly in the skeleton by mesenchymal stromal cells (MSC) and MSC-derived osteoblasts. Histochemical analysis of human multiple myeloma specimens also demonstrated that MMP13 largely localizes to the stromal compartment compared with CD138+ myeloma cells. In this study, we further identified that multiple myeloma induces MMP13 expression in bone stromal cells. Because of its ability to degrade type I collagen, we examined whether bone stromal–derived MMP13 contributed to myeloma progression. Multiple myeloma cells were inoculated into wild-type or MMP13–null mice. In independent in vivo studies, MMP13–null mice demonstrated significantly higher overall survival rates and lower levels of bone destruction compared with wild-type controls. Unexpectedly, no differences in type I collagen processing between the groups were observed. Ex vivo stromal coculture assays showed reduced formation and activity in MMP13–null osteoclasts. Analysis of soluble factors from wild-type and MMP13–null MSCs revealed decreased bioavailability of various osteoclastogenic factors including CXCL7. CXCL7 was identified as a novel MMP13 substrate and regulator of osteoclastogenesis. Underscoring the importance of host MMP13 catalytic activity in multiple myeloma progression, we demonstrate the in vivo efficacy of a novel and highly selective MMP13 inhibitor that provides a translational opportunity for the treatment of this incurable disease.Significance:Genetic and pharmacologic approaches show that bone stromal–derived MMP13 catalytic activity is critical for osteoclastogenesis, bone destruction, and disease progression.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Data from Host-Derived Matrix Metalloproteinase-13 Activity Promotes Multiple Myeloma–Induced Osteolysis and Reduces Overall Survival
- Author
-
Conor C. Lynch, Gregg B. Fields, Anna M. Knapinska, William R. Roush, Rita Fuerst, Jun Yong Choi, Kenneth H. Shain, Tao Li, Jeremy J. McGuire, Gemma Shay, and Chen Hao Lo
- Abstract
All Supplementary Figures and Legends
- Published
- 2023
- Full Text
- View/download PDF
11. Phagocytosis increases an oxidative metabolic and immune suppressive signature in tumor macrophages
- Author
-
Michael A. Gonzalez, Daniel R. Lu, Maryam Yousefi, Ashley Kroll, Chen Hao Lo, Carlos G. Briseño, J. E. Vivienne Watson, Sergey Novitskiy, Vanessa Arias, Hong Zhou, Andres Plata Stapper, Min K. Tsai, Emily L. Ashkin, Christopher W. Murray, Chi-Ming Li, Monte M. Winslow, and Kristin V. Tarbell
- Subjects
Immunology ,Immunology and Allergy - Abstract
Phagocytosis is a key macrophage function, but how phagocytosis shapes tumor-associated macrophage (TAM) phenotypes and heterogeneity in solid tumors remains unclear. Here, we utilized both syngeneic and novel autochthonous lung tumor models in which neoplastic cells express the fluorophore tdTomato (tdTom) to identify TAMs that have phagocytosed neoplastic cells in vivo. Phagocytic tdTompos TAMs upregulated antigen presentation and anti-inflammatory proteins, but downregulated classic proinflammatory effectors compared to tdTomneg TAMs. Single-cell transcriptomic profiling identified TAM subset-specific and common gene expression changes associated with phagocytosis. We uncover a phagocytic signature that is predominated by oxidative phosphorylation (OXPHOS), ribosomal, and metabolic genes, and this signature correlates with worse clinical outcome in human lung cancer. Expression of OXPHOS proteins, mitochondrial content, and functional utilization of OXPHOS were increased in tdTompos TAMs. tdTompos tumor dendritic cells also display similar metabolic changes. Our identification of phagocytic TAMs as a distinct myeloid cell state links phagocytosis of neoplastic cells in vivo with OXPHOS and tumor-promoting phenotypes.
- Published
- 2023
- Full Text
- View/download PDF
12. Computational modeling reveals a key role for polarized myeloid cells in controlling osteoclast activity during bone injury repair
- Author
-
Conor C. Lynch, David Basanta, Etienne Baratchart, and Chen Hao Lo
- Subjects
0301 basic medicine ,Male ,Differential equations ,musculoskeletal diseases ,Cell type ,Myeloid ,Bone Regeneration ,Population dynamics ,Osteoimmunology ,Science ,Population ,Osteoclasts ,Bone healing ,Models, Biological ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Osteoclast ,Dynamical systems ,medicine ,Animals ,Computational models ,Computer Simulation ,education ,Bone ,education.field_of_study ,Multidisciplinary ,Tibia ,Chemistry ,Bone Injury ,Osteoblast ,Cell biology ,Resorption ,Innate immune cells ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Computer modelling ,Multicellular systems ,Medicine ,Data integration ,Bone marrow - Abstract
Bone-forming osteoblasts and -resorbing osteoclasts control bone injury repair, and myeloid-derived cells such as monocytes and macrophages are known to influence their behavior. However, precisely how these multiple cell types coordinate and regulate each other over time within the bone marrow to restore bone is difficult to dissect using biological approaches. Conversely, mathematical modeling lends itself well to this challenge. Therefore, we generated an ordinary differential equation (ODE) model powered by experimental data (osteoblast, osteoclast, bone volume, pro- and anti-inflammatory myeloid cells) obtained from intra-tibially injured mice. Initial ODE results using only osteoblast/osteoclast populations demonstrated that bone homeostasis could not be recovered after injury, but this issue was resolved upon integration of pro- and anti-inflammatory myeloid population dynamics. Surprisingly, the ODE revealed temporal disconnects between the peak of total bone mineralization/resorption, and osteoblast/osteoclast numbers. Specifically, the model indicated that osteoclast activity must vary greatly (> 17-fold) to return the bone volume to baseline after injury and suggest that osteoblast/osteoclast number alone is insufficient to predict bone the trajectory of bone repair. Importantly, the values of osteoclast activity fall within those published previously. These data underscore the value of mathematical modeling approaches to understand and reveal new insights into complex biological processes.
- Published
- 2021
13. Host-Derived Matrix Metalloproteinase-13 Activity Promotes Multiple Myeloma–Induced Osteolysis and Reduces Overall Survival
- Author
-
Conor C. Lynch, Kenneth H. Shain, William R. Roush, Tao Li, Gemma Shay, Anna M. Knapinska, Gregg B. Fields, Chen Hao Lo, Jeremy J. McGuire, Jun Yong Choi, and Rita Fuerst
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Stromal cell ,Osteolysis ,Osteoclasts ,Mice, Transgenic ,Matrix metalloproteinase ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Osteogenesis ,In vivo ,Cell Line, Tumor ,Matrix Metalloproteinase 13 ,medicine ,Animals ,Humans ,Multiple myeloma ,Osteoblasts ,Chemistry ,Mesenchymal stem cell ,Wild type ,Cell Differentiation ,Mesenchymal Stem Cells ,medicine.disease ,Mice, Inbred C57BL ,Survival Rate ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Female ,Multiple Myeloma ,Chemokines, CXC ,Ex vivo ,Signal Transduction - Abstract
Multiple myeloma promotes systemic skeletal bone disease that greatly contributes to patient morbidity. Resorption of type I collagen–rich bone matrix by activated osteoclasts results in the release of sequestered growth factors that can drive progression of the disease. Matrix metalloproteinase-13 (MMP13) is a collagenase expressed predominantly in the skeleton by mesenchymal stromal cells (MSC) and MSC-derived osteoblasts. Histochemical analysis of human multiple myeloma specimens also demonstrated that MMP13 largely localizes to the stromal compartment compared with CD138+ myeloma cells. In this study, we further identified that multiple myeloma induces MMP13 expression in bone stromal cells. Because of its ability to degrade type I collagen, we examined whether bone stromal–derived MMP13 contributed to myeloma progression. Multiple myeloma cells were inoculated into wild-type or MMP13–null mice. In independent in vivo studies, MMP13–null mice demonstrated significantly higher overall survival rates and lower levels of bone destruction compared with wild-type controls. Unexpectedly, no differences in type I collagen processing between the groups were observed. Ex vivo stromal coculture assays showed reduced formation and activity in MMP13–null osteoclasts. Analysis of soluble factors from wild-type and MMP13–null MSCs revealed decreased bioavailability of various osteoclastogenic factors including CXCL7. CXCL7 was identified as a novel MMP13 substrate and regulator of osteoclastogenesis. Underscoring the importance of host MMP13 catalytic activity in multiple myeloma progression, we demonstrate the in vivo efficacy of a novel and highly selective MMP13 inhibitor that provides a translational opportunity for the treatment of this incurable disease.Significance:Genetic and pharmacologic approaches show that bone stromal–derived MMP13 catalytic activity is critical for osteoclastogenesis, bone destruction, and disease progression.
- Published
- 2021
- Full Text
- View/download PDF
14. Integrated computational and in vivo models reveal Key Insights into Macrophage Behavior during bone healing
- Author
-
Etienne Baratchart, Chen Hao Lo, Conor C. Lynch, and David Basanta
- Abstract
Myeloid-derived monocyte and macrophages are key cells in the bone that contribute to remodeling and injury repair. However, their temporal polarization status and control of boneresorbing osteoclasts and bone-forming osteoblasts responses is largely unknown. In this study, we focused on two aspects of monocyte/macrophage dynamics and polarization states over time: 1) the injury-triggered pro- and anti-inflammatory monocytes/macrophages temporal profiles, 2) the contributions of pro-versus anti-inflammatory monocytes/macrophages in coordinating healing response. Bone healing is a complex multicellular dynamic process. While traditional in vitro and in vivo experimentation may capture the behavior of select populations with high resolution, they cannot simultaneously track the behavior of multiple populations. To address this, we have used an integrated a coupled ordinary differential equations (ODEs)-based framework describing multiple cellular species to in vivo bone injury data in order to identify and test various hypotheses regarding bone cell populations dynamics. Our approach allowed us to infer several biological insights including, but not limited to,: 1) anti-inflammatory macrophages are key for early osteoclast inhibition and pro-inflammatory macrophage suppression, 2) pro-inflammatory macrophages are involved in osteoclast bone resorptive activity, whereas osteoblasts promote osteoclast differentiation, 3) Pro-inflammatory monocytes/macrophages rise during two expansion waves, which can be explained by the anti-inflammatory macrophages-mediated inhibition phase between the two waves. In addition, we further tested the robustness of the mathematical model by comparing simulation results to an independent experimental dataset. Taken together, this novel comprehensive mathematical framework allowed us to identify biological mechanisms that best recapitulate bone injury data and that explain the coupled cellular population dynamics involved in the process. Furthermore, our hypothesis testing methodology could be used in other contexts to decipher mechanisms in complex multicellular processes.Author SummaryMyeloid-derived monocytes/macrophages are key cells for bone remodeling and injury repair. However, their temporal polarization status and control of bone-resorbing osteoclasts and boneforming osteoblasts responses is largely unknown. In this study, we focused on two aspects of monocyte/macrophage population dynamics: 1) the injury-triggered pro- and anti-inflammatory monocytes/macrophages temporal profiles, 2) the contributions of pro-versus anti-inflammatory monocytes/macrophages in coordinating healing response. In order to test various hypotheses regarding bone cell populations dynamics, we have integrated a coupled ordinary differential equations-based framework describing multiple cellular species to in vivo bone injury data. Our approach allowed us to infer several biological insights including: 1) anti-inflammatory macrophages are key for early osteoclast inhibition and pro-inflammatory macrophage suppression, 2) pro-inflammatory macrophages are involved in osteoclast bone resorptive activity, whereas osteoblasts promote osteoclast differentiation, 3) Pro-inflammatory monocytes/macrophages rise during two expansion waves, which can be explained by the anti-inflammatory macrophages-mediated inhibition phase between the two waves. Taken together, this mathematical framework allowed us to identify biological mechanisms that recapitulate bone injury data and that explain the coupled cellular population dynamics involved in the process.
- Published
- 2021
- Full Text
- View/download PDF
15. Mesenchymal stem cell-derived interleukin-28 drives the selection of apoptosis resistant bone metastatic prostate cancer
- Author
-
Harshani R. Lawrence, Leah M. Cook, Conor C. Lynch, Jeremy J. McGuire, Tao Li, Jeremy S. Frieling, Nicholas J. Lawrence, Chen Hao Lo, and Ayaz Muhammad
- Subjects
0301 basic medicine ,Male ,Cell ,General Physics and Astronomy ,Apoptosis ,Docetaxel ,Metastasis ,Prostate cancer ,0302 clinical medicine ,Bone cancer ,RNA, Small Interfering ,STAT3 ,Receptors, Interferon ,Mice, Knockout ,Multidisciplinary ,biology ,Benzenesulfonates ,Cell Differentiation ,Recombinant Proteins ,medicine.anatomical_structure ,STAT1 Transcription Factor ,030220 oncology & carcinogenesis ,medicine.drug ,Cancer microenvironment ,STAT3 Transcription Factor ,Science ,Primary Cell Culture ,Bone Neoplasms ,Urological cancer ,Adenocarcinoma ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cell Proliferation ,Osteoblasts ,Tibia ,business.industry ,Cell growth ,Mesenchymal stem cell ,Prostatic Neoplasms ,Mesenchymal Stem Cells ,General Chemistry ,medicine.disease ,Aminosalicylic Acids ,Disease Models, Animal ,030104 developmental biology ,Culture Media, Conditioned ,biology.protein ,Cancer research ,Bone marrow ,Interferons ,business - Abstract
Bone metastatic prostate cancer (PCa) promotes mesenchymal stem cell (MSC) recruitment and their differentiation into osteoblasts. However, the effects of bone-marrow derived MSCs on PCa cells are less explored. Here, we report MSC-derived interleukin-28 (IL-28) triggers prostate cancer cell apoptosis via IL-28 receptor alpha (IL-28Rα)-STAT1 signaling. However, chronic exposure to MSCs drives the selection of prostate cancer cells that are resistant to IL-28-induced apoptosis and therapeutics such as docetaxel. Further, MSC-selected/IL-28-resistant prostate cancer cells grow at accelerated rates in bone. Acquired resistance to apoptosis is PCa cell intrinsic, and is associated with a shift in IL-28Rα signaling via STAT1 to STAT3. Notably, STAT3 ablation or inhibition impairs MSC-selected prostate cancer cell growth and survival. Thus, bone marrow MSCs drive the emergence of therapy-resistant bone metastatic prostate cancer yet this can be disabled by targeting STAT3., The effects of bone-marrow derived MSCs on prostate cancer cells remain unknown. Here the authors show that MSC-derived IL-28 induces prostate cancer cell apoptosis via IL-28Rα-STAT1 signalling, while acquired resistance to apoptosis is associated with a shift in IL-28Rα signalling via STAT1 to STAT3.
- Published
- 2021
16. Autocrine TGFβ Is a Survival Factor for Monocytes and Drives Immunosuppressive Lineage Commitment
- Author
-
Alba Gonzalez-Junca, Shisuo Du, Iliana Tenvooren, Diana M. Marquez, Renate Parry, Mary Helen Barcellos-Hoff, Ritu Roy, Matthew H. Spitzer, Ilenia Pellicciotta, Chen Hao Lo, and Kyla Driscoll
- Subjects
Cancer Research ,Myeloid ,Cell Survival ,CD14 ,Immunology ,Oncology and Carcinogenesis ,Adenocarcinoma of Lung ,Biology ,Article ,Monocytes ,Transforming Growth Factor beta1 ,Immunomodulation ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Transforming Growth Factor beta ,medicine ,Humans ,Autocrine signalling ,Tumor microenvironment ,Antigen Presentation ,Gene Expression Profiling ,Myeloid-Derived Suppressor Cells ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,Pharmacology and Pharmaceutical Sciences ,Colony-stimulating factor ,Autocrine Communication ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Biomarkers ,030215 immunology ,Transforming growth factor ,Signal Transduction - Abstract
Transforming growth factor β (TGFβ) is an effector of immune suppression and contributes to a permissive tumor microenvironment that compromises effective immunotherapy. We identified a correlation between TGFB1 and genes expressed by myeloid cells, but not granulocytes, in The Cancer Genome Atlas lung adenocarcinoma data, in which high TGFB1 expression was associated with poor survival. To determine whether TGFβ affected cell fate decisions and lineage commitment, we studied primary cultures of CD14+ monocytes isolated from peripheral blood of healthy donors. We discovered that TGFβ was a survival factor for CD14+ monocytes, which rapidly executed an apoptotic program in its absence. Continued exposure to TGFβ in combination with granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin 6 (IL6) amplified HLA-DRlowCD14+CD11b+CD33+ myeloid-derived suppressor cells (MDSCs) at the expense of macrophage and dendritic cell (DC) differentiation. MDSCs generated in the presence of TGFβ were more effective in suppressing T-cell proliferation and promoted the T regulatory cell phenotype. In contrast, inhibition of TGFβ signaling using a small-molecule inhibitor of receptor kinase activity in CD14+ monocytes treated with GM-CSF and IL6 decreased MDSC differentiation and increased differentiation to proinflammatory macrophages and antigen-presenting DCs. The effect of autocrine and paracrine TGFβ on myeloid cell survival and lineage commitment suggests that pharmacologic inhibition of TGFβ-dependent signaling in cancer would favor antitumor immunity.
- Published
- 2019
- Full Text
- View/download PDF
17. Arginine vasopressin receptor 1a is a therapeutic target for castration-resistant prostate cancer
- Author
-
Laine Heidman, Fiorella Magani, Kerry L. Burnstein, Chen Hao Lo, Meghan A. Rice, Yushan Zhang, Maria J. Martinez, Stephanie Peacock, Yehia Daaka, Conor C. Lynch, Cale D. Fahrenholtz, Valeria A. Copello, Ann M. Greene, and Ning Zhao
- Subjects
MAPK/ERK pathway ,Male ,Receptors, Vasopressin ,desmopressin ,Indoles ,Pyrrolidines ,medicine.drug_class ,MAP Kinase Signaling System ,vasopressin ,Mice, Nude ,urologic and male genital diseases ,relcovaptan ,Article ,Androgen deprivation therapy ,Prostate cancer ,Castration Resistance ,Osteogenesis ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Molecular Targeted Therapy ,RNA, Messenger ,Receptor ,Cyclic AMP Response Element-Binding Protein ,Proto-Oncogene Proteins c-vav ,Cell Proliferation ,Arginine vasopressin receptor 1A ,drug repurposing ,business.industry ,General Commentary ,General Medicine ,Androgen ,medicine.disease ,hormone-resistant cancer ,Androgen receptor ,Gene Expression Regulation, Neoplastic ,Prostatic Neoplasms, Castration-Resistant ,Oncology ,Drug Resistance, Neoplasm ,Receptors, Androgen ,Cancer research ,Calcium ,business - Abstract
Castration-resistant prostate cancer (CRPC) recurs after androgen deprivation therapy (ADT) and is incurable. Reactivation of androgen receptor (AR) signaling in the low androgen environment of ADT drives CRPC. This AR activity occurs through a variety of mechanisms, including up-regulation of AR coactivators such as VAV3 and expression of constitutively active AR variants such as the clinically relevant AR-V7. AR-V7 lacks a ligand-binding domain and is linked to poor prognosis. We previously showed that VAV3 enhances AR-V7 activity to drive CRPC progression. Gene expression profiling after depletion of either VAV3 or AR-V7 in CRPC cells revealed arginine vasopressin receptor 1a (AVPR1A) as the most commonly down-regulated gene, indicating that this G protein–coupled receptor may be critical for CRPC. Analysis of publicly available human PC datasets showed that AVPR1A has a higher copy number and increased amounts of mRNA in advanced PC. Depletion of AVPR1A in CRPC cells resulted in decreased cell proliferation and reduced cyclin A. In contrast, androgen-dependent PC, AR-negative PC, or nontumorigenic prostate epithelial cells, which have undetectable AVPR1A mRNA, were minimally affected by AVPR1A depletion. Ectopic expression of AVPR1A in androgen-dependent PC cells conferred castration resistance in vitro and in vivo. Furthermore, treatment of CRPC cells with the AVPR1A ligand, arginine vasopressin (AVP), activated ERK and CREB, known promoters of PC progression. A clinically safe and selective AVPR1A antagonist, relcovaptan, prevented CRPC emergence and decreased CRPC orthotopic and bone metastatic growth in mouse models. Based on these preclinical findings, repurposing AVPR1A antagonists is a promising therapeutic approach for CRPC.
- Published
- 2018
18. Abstract 934: γδ CAR T-cell therapy significantly mitigates bone metastatic castrate-resistant prostate cancer
- Author
-
Jeremy S. Frieling, Chen Hao Lo, Ismahene Benzaid, Conor C. Lynch, María Cecilia Ramello, Daniel Abate-Daga, and Emiliano Roselli
- Subjects
Cancer Research ,Bone disease ,business.industry ,medicine.medical_treatment ,Bone metastasis ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Prostate cancer ,Oncology ,Antigen ,Cancer research ,medicine ,Stem cell ,business ,Homing (hematopoietic) - Abstract
Bone metastasis is a frequent complication in advanced prostate cancer, with the resultant lesions significantly contributing to patient morbidity and mortality. While next generation hormone ablation therapies and bone protecting bisphosphonates alleviate these symptoms, the disease remains incurable, and new therapeutic approaches are of urgent need. In this regard, we have focused on tapping the tumor-seeking potential of chimeric antigen receptor (CAR) T cells. CAR T cells have shown remarkable anti-tumor responses in hematologic malignancies, but unique physical and biological challenges have hindered their activity in solid tumors. Interestingly, patients treated with bisphosphonates such as zoledronate exhibit enhanced recruitment and activation of the γδ subset of T cells in bone due intracellular accumulation of isopentenyl pyrophosphate (IPP) phosphoantigen. Whereas conventional CAR T cell therapies utilize αβ T cells, we posit that designing γδ CAR T cells could improve homing to bone metastases when administered with bisphosphonates. First, to test the impact of prostate specific stem cell antigen (PSCA)-specific γδ CAR T cells against bone metastatic prostate cancer in vivo, NSG mice (n=10) were intratibially injected with PSCA/luciferase-expressing C4-2B (2x105) castrate resistant prostate cancer cells. Tumors were allowed to establish for 10 days and then randomized into control or γδ CAR T (1.5x107 via tail vein) groups. Subsequent bioluminescent imaging indicated a rapid and significant (p=0.0006) regression of tumors in the γδ CAR T cell group, leading to increased overall survival (5/5 γδ CAR T vs. 0/5 control after 68 days, p=0.0002). Ex vivo bone morphometry analysis also demonstrated the significant protective effect of γδ CAR T associated bone disease. To determine whether bisphosphonates could further enhance the homing of γδ CAR T to bone, NSG mice (n=30) were intratibially injected with C4-2B (2x105), and randomized into control and zoledronate (30µg/kg) groups. After 10 days, mice received γδ T cells (3x106). Subsets were sacrificed at 1, 3, and 5 days post-T cell administration, and peripheral blood, tibia bone marrow, and spleens isolated. CD3-Vδ2 flow cytometry indicated increased γδ T cells in the tibia bone marrow from zoledronate groups (Day 1=61%, Day 3=32%, and Day 5=57%). Furthermore, decreased tumor growth rates were observed in the zoledronate group, suggesting that increased homing of γδ T cells induced anti-tumor effects. Our data to date demonstrate that γδ CAR T cells significantly mitigate bone metastatic prostate cancer and associated bone disease. Further, bisphosphonates (already used in the clinical setting) enhance the homing of γδ CAR T cells to the tumor-bone microenvironment. We posit that γδ CAR T will be an effective immunotherapy approach for the treatment of men with incurable bone metastatic prostate cancer. Citation Format: Jeremy Steven Frieling, Maria Cecilia Ramello, Ismahene Benzaid, Emiliano Roselli, Chen Hao Lo, Conor C. Lynch, Daniel Abate-Daga. γδ CAR T-cell therapy significantly mitigates bone metastatic castrate-resistant prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 934.
- Published
- 2019
- Full Text
- View/download PDF
19. Host-derived MMP-13 promotes Multiple Myeloma skeletal colonization
- Author
-
Conor C. Lynch, Lori A. Hazlehurst, Chen Hao Lo, and Gemma Shay
- Subjects
Host (biology) ,medicine ,Cancer research ,Colonization ,General Medicine ,Biology ,Matrix metalloproteinase ,medicine.disease ,Multiple myeloma - Published
- 2016
- Full Text
- View/download PDF
20. Abstract A063: Preclinical evaluation of an arginine vasopressin receptor 1A (AVPR1A) antagonist in castration-resistant prostate cancer
- Author
-
Stephanie Peacock, Yushan Zhang, Meghan A. Rice, Ann M. Greene, Ning Zhao, Fiorella Magani, Laine Heidman, Chen Hao Lo, Yehia Daaka, Conor C. Lynch, and Kerry L. Burnstein
- Subjects
Cancer Research ,business.industry ,Cancer ,urologic and male genital diseases ,medicine.disease ,Androgen receptor ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Castration Resistance ,Downregulation and upregulation ,Tumor progression ,Prostate ,LNCaP ,medicine ,Cancer research ,business - Abstract
Background: Advanced prostate cancer is treated by androgen deprivation, but despite initial responses most tumors inevitably recur. The recurrent disease is termed castration-resistant prostate cancer (CRPC) and is characterized by active androgen receptor (AR) signaling despite decreased androgen levels. AR activity in CRPC occurs through a variety of mechanisms including upregulation of AR coactivators, such as Vav3, and expression of constitutively active AR variants, which lack the ligand binding domain and are linked to poor prognosis. We previously demonstrated that PC cells are growth inhibited by depletion of either Vav3 or the clinically prevalent AR variant, AR-V7 (1). Gene expression profiling in CRPC cells revealed that arginine vasopressin receptor 1a (AVPR1a) was significantly reduced by depletion of either Vav3 or AR-V7. AVPR1a is a G protein-coupled receptor that has not been previously linked to PC. Analysis of publicly available datasets showed that the AVPR1a gene exhibits a significant increase in copy number in human CRPC specimens and AVPR1a mRNA is upregulated in aggressive PC (2-4). Methods: To explore the role of AVPR1a in PC cells, we selectively depleted AVPR1a in a panel of prostate cancer and prostate epithelial cell lines and examined cell survival and proliferation in culture and soft agar, as well as cell migration and invasion. Conversely, we overexpressed AVPR1a in an androgen-dependent PC cell line, LNCaP, and examined growth in vitro and in vivo in androgen-depleted conditions. We investigated AVPR1a antagonism in two distinct in vivo models of CRPC progression and growth using an effective and safe-in-humans AVPR1a antagonist, relcovaptan. Results: We showed that depletion of AVPR1a greatly inhibited the proliferation of multiple CRPC cell lines but had less to no effect on androgen-dependent PC cells or nontumorigenic prostate epithelial cells. Depletion of AVPR1a also decreased CRPC cell anchorage-independent growth. Conversely, overexpression of AVPR1a in androgen-dependent LNCaP cells conferred castration resistance in vitro and in vivo. Similar to depletion of AVPR1a, treatment of PC cells with the AVPR1a antagonist relcovaptan decreased CRPC anchorage-independent growth. AVPR1a depletion or antagonism was also effective at decreasing CRPC metastatic properties: migration and invasion. In a preclinical xenograft model of progression to castration resistance, we found that relcovaptan halted tumor growth and stabilized circulating levels of a clinical tumor progression marker, prostate specific antigen (PSA). We evaluated relcovaptan efficacy on end-stage metastatic CRPC growth. C4-2B CRPC cells were injected into the tibias of castrated mice to model tumor growth and bone remodeling. Relcovaptan significantly inhibited tumor growth and prevented cancer cell-induced bone remodeling. Conclusion: AVPR1a is a promising new therapeutic target against CRPC. We propose that AVPR1a antagonists may be repurposed for PC therapy. References: 1. Peacock SO, et al. Mol Endocrinol 2012;12:1967-79. 2. Chandran UR, et al. BMC Cancer 2007;7:64. 3. Tamura K, et al. BMC Cancer 2007;67:5117-25. 4. Beltran H, et al. Nat Med 2016;22;298-305. Citation Format: Ning Zhao, Stephanie Peacock, Chen Hao Lo, Laine Heidman, Fiorella Magani, Meghan Rice, Ann Greene, Yushan Zhang, Yehia Daaka, Conor Lynch, Kerry Burnstein. Preclinical evaluation of an arginine vasopressin receptor 1A (AVPR1A) antagonist in castration-resistant prostate cancer [abstract]. In: Proceedings of the AACR Special Conference: Prostate Cancer: Advances in Basic, Translational, and Clinical Research; 2017 Dec 2-5; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(16 Suppl):Abstract nr A063.
- Published
- 2018
- Full Text
- View/download PDF
21. Abstract 4260: Dynamic modeling of macrophage plasticity in bone metastatic prostate cancer
- Author
-
Etienne Baratchart, Conor C. Lynch, Chen Hao Lo, and David Basanta
- Subjects
Cancer Research ,Stromal cell ,business.industry ,Bone Injury ,Macrophage polarization ,Osteoblast ,Bone healing ,medicine.disease ,Bone remodeling ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Osteoclast ,medicine ,Cancer research ,business - Abstract
Tumor-associated macrophages, particularly those with an anti-inflammatory phenotype, have long been implicated in the progression of primary solid malignancies, including prostate cancer. Metastatic prostate cancer typically manifests in the bone where it induces painful osteogenic lesions that are incurable. Bone is naturally rich in myeloid-derived macrophages whose temporal polarization into pro- (M1) and anti-inflammatory (M2) phenotypes is critical for regulating the bone repair program mediated by bone-resorbing osteoclasts and bone-building osteoblasts. However, the dynamics of macrophage polarization in the context of bone metastatic prostate cancer are underexplored and difficult to address with traditional biologic approaches. To address the role of macrophage polarization in the context of bone metastatic prostate cancer, we first investigated the macrophage polarization temporal dynamics in normal bones and analyzed macrophage plasticity in vivo subsequent to intratibial injury. Bone marrows were isolated at several time points and profiled by flow cytometry for pro- and anti-inflammatory monocyte macrophage content. Contralateral tibias were analyzed for bone volume, osteoblast and osteoclast numbers. We then designed a mathematical model describing the different cell population dynamics in the bones. Generation of the model required testing a number of assumptions regarding macrophage polarization behavior. For example, it is unknown whether M1 resolves at the initial stages of bone injury repair through death or by repolarizing into M2. For each aspect, competing mechanistic assumptions were proposed and simulated by sets of ODEs. The best-fitting assumptions for each aspect were integrated into a single comprehensive ODE model to fully describe the dynamics of the bone resident cell populations during bone remodeling. This experimentally validated model is now being used to address how bone resident cells respond to metastatic prostate cancer cells. In conclusion, we have generated an ODE model that describes macrophage polarization over time during bone repair and how pro- and anti-inflammatory macrophages interact with bone stromal cells. The mathematical model predictions are in agreement with our biologic experiments in vivo and will allow us to interrogate how macrophage polarization impacts the behavior of metastatic prostate cancer cells in the bone microenvironment. Citation Format: Etienne A. Baratchart, Chen Hao Lo, Conor Lynch, David Basanta. Dynamic modeling of macrophage plasticity in bone metastatic prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4260.
- Published
- 2018
- Full Text
- View/download PDF
22. Attenuation of the DNA damage response by transforming growth factor-beta inhibitors enhances radiation sensitivity of non-small-cell lung cancer cells in vitro and in vivo
- Author
-
S.F. Bouquet, Ilenia Pellicciotta, Mary Helen Barcellos-Hoff, Shiva Bolourchi, Renate Parry, Chen Hao Lo, and Shisuo Du
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,DNA damage ,Blotting, Western ,Receptor, Transforming Growth Factor-beta Type I ,Ataxia Telangiectasia Mutated Proteins ,In Vitro Techniques ,Protein Serine-Threonine Kinases ,Radiation Tolerance ,Histones ,Mice ,Radiation sensitivity ,Transforming Growth Factor beta ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Radiosensitivity ,Amino Acids ,Phosphorylation ,Clonogenic assay ,A549 cell ,Radiation ,biology ,business.industry ,Antibodies, Monoclonal ,Transforming growth factor beta ,Antibodies, Neutralizing ,Comet assay ,Oncology ,Xanthenes ,Chemotherapy, Adjuvant ,Cancer research ,biology.protein ,Comet Assay ,Tumor Suppressor Protein p53 ,business ,Receptors, Transforming Growth Factor beta ,Transforming growth factor ,DNA Damage - Abstract
Purpose To determine whether transforming growth factor (TGF)-β inhibition increases the response to radiation therapy in human and mouse non–small-cell lung carcinoma (NSCLC) cells in vitro and in vivo. Methods and Materials TGF-β–mediated growth response and pathway activation were examined in human NSCLC NCI-H1299, NCI-H292, and A549 cell lines and murine Lewis lung cancer (LLC) cells. Cells were treated in vitro with LY364947, a small-molecule inhibitor of the TGF-β type 1 receptor kinase, or with the pan-isoform TGF-β neutralizing monoclonal antibody 1D11 before radiation exposure. The DNA damage response was assessed by ataxia telangiectasia mutated (ATM) or Trp53 protein phosphorylation, γH2AX foci formation, or comet assay in irradiated cells. Radiation sensitivity was determined by clonogenic assay. Mice bearing syngeneic subcutaneous LLC tumors were treated with 5 fractions of 6 Gy and/or neutralizing or control antibody. Results The NCI-H1299, A549, and LLC NSCLC cell lines pretreated with LY364947 before radiation exposure exhibited compromised DNA damage response, indicated by decreased ATM and p53 phosphorylation, reduced γH2AX foci, and increased radiosensitivity. The NCI-H292 cells were unresponsive. Transforming growth factor-β signaling inhibition in irradiated LLC cells resulted in unresolved DNA damage. Subcutaneous LLC tumors in mice treated with TGF-β neutralizing antibody exhibited fewer γH2AX foci after irradiation and significantly greater tumor growth delay in combination with fractionated radiation. Conclusions Inhibition of TGF-β before radiation attenuated DNA damage recognition and increased radiosensitivity in most NSCLC cells in vitro and promoted radiation-induced tumor control in vivo. These data support the rationale for concurrent TGF-β inhibition and RT to provide therapeutic benefit in NSCLC.
- Published
- 2014
23. Abstract 3282: Host-derived MMP-13 mediates multiple myeloma-induced osteolysis
- Author
-
Conor C. Lynch, Chen Hao Lo, and Gemma Shay
- Subjects
Cancer Research ,Osteolysis ,business.industry ,Mesenchymal stem cell ,Cancer ,medicine.disease ,Bone remodeling ,Oncology ,In vivo ,Cathepsin K ,medicine ,Cancer research ,business ,Multiple myeloma ,Ex vivo - Abstract
Myeloma cells promote osteolysis and suppress osteogenesis in the bone microenvironment by regulating osteoclasts (OCL) and osteoblasts (OBL), respectively. Patients often suffer painful, osteoporotic bone status as diseases progresses. Bone consists of 90% type-I collagen, which is degraded by OCL derived cathepsin K during regular bone remodeling. While bisphosphonates delay pathological fracture, they have little impact on overall survival; therefore, development of new therapies is crucial. Matrix metalloproteinase-13 (MMP-13) is an enzyme involved in type-I collagen degradation and resorption. Immunohistochemical staining of a panel of human myeloma biopsies (n = 15) show high MMP-13 expression predominantly in bone stroma and to a lesser extent in the myeloma compartment. Consistent with previous reports, MMP-13 expression localizes to mesenchymal stromal cells (MSC) and OBLs rather than OCLs. Further supporting these observations, analysis of publically available data sets revealed upregulation of MMP-13 expression in MSCs co-cultured with myeloma cells (1.81 LogFC, p To test our hypothesis, we generated immune-compromised MMP-13-null mice on a C57BL/6 background for ex vivo and in vivo studies. Mouse myeloma cell line, 5TGM1, homes to the skeleton and progresses upon tail vein injection in this model. Our in vivo studies showed that 5TGM1-bearing MMP-13-null mice had significantly improved overall survival compared to wild-type controls (Mean 39 vs. 43 days; p Citation Format: Chen Hao Lo, Gemma Shay, Conor C. Lynch. Host-derived MMP-13 mediates multiple myeloma-induced osteolysis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3282.
- Published
- 2016
- Full Text
- View/download PDF
24. γδ-Enriched CAR-T cell therapy for bone metastatic castrate-resistant prostate cancer.
- Author
-
Frieling, Jeremy S., Tordesillas, Leticia, Bustos, Xiomar E., Ramello, Maria Cecilia, Bishop, Ryan T., Cianne, Junior E., Snedal, Sebastian A., Tao Li, Chen Hao Lo, de la Iglesia, Janis, Roselli, Emiliano, Benzaïd, Ismahène, Xuefeng Wang, Youngchul Kim, Lynch, Conor C., and Abate-Daga, Daniel
- Abstract
The article focuses on the use of γδ-enriched CAR-T cell therapy in the treatment of bone metastatic castrate-resistant prostate cancer (mCRPC). The topics include the effectiveness of γδ CAR-T cells targeting prostate stem cell antigen (PSCA) in regressing tumors, the role of zoledronate (ZOL) in enhancing γδ CAR-T cell activation and antitumor efficacy, and the potential of γδ CAR-T cell therapy for mCRPC treatment.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.