9 results on '"Mitchell, Joylise A."'
Search Results
2. Enhanced tumor control and survival in preclinical models with adoptive cell therapy preceded by low-dose radiotherapy.
- Author
-
Puebla-Osorio, Nahum, Fowlkes, Natalie Wall, Barsoumian, Hampartsoum B., Xega, Kristina, Srivastava, Gitika, Kettlun-Leyton, Claudia, Nizzero, Sara, Voss, Tiffany, Riad, Thomas S., Wong, Christina, Huang, Ailing, Yun Hu, Mitchell, Joylise, Mingee Kim, Rafiq, Zahid, Kewen He, Sezen, Duygu, Hsu, Ethan, Masrorpour, Fatemeh, and Maleki, Aurian
- Subjects
CHIMERIC antigen receptors ,ATOMIC force microscopy ,TREATMENT effectiveness ,TUMOR growth ,SURVIVAL rate - Abstract
Introduction: Effective infiltration of chimeric antigen receptor T (CAR-T) cells into solid tumors is critical for achieving a robust antitumor response and improving therapeutic outcomes. While CAR-T cell therapies have succeeded in hematologic malignancies, their efficacy in solid tumors remains limited due to poor tumor penetration and an immunosuppressive tumor microenvironment. This study aimed to evaluate the potential of low-dose radiotherapy (LDRT) administered before T-cell therapy to enhance the antitumor effect by promoting CAR-T cell infiltration. We hypothesized that combining LDRT with T-cell therapy would improve tumor control and survival compared to either treatment alone. Methods: We investigated this hypothesis using two NSG mouse models bearing GSU or CAPAN-2 solid tumors. The mice were treated with engineered CAR-T cells targeting guanyl cyclase-C (GCC) or mesothelin as monotherapy or in combination with LDRT. Additionally, we extended this approach to a C57BL/6 mouse model implanted with MC38-gp100+ cells, followed by adoptive transfer of pmel+ T cells before and after LDRT. Tumor growth and survival outcomes were monitored in all models. Furthermore, we employed atomic force microscopy (AFM) in a small cohort to assess the effects of radiotherapy on tumor stiffness and plasticity, exploring the role of tumor nanomechanics as a potential biomarker for treatment efficacy. Results: Our results demonstrated enhanced tumor control and prolonged survival in mice treated with LDRT followed by T-cell therapy across all models. The combination of LDRT with CAR-T or pmel+ T-cell therapy led to superior tumor suppression and survival compared to monotherapy, highlighting the synergistic impact of the combined approach. Additionally, AFM analysis revealed significant changes in tumor stiffness and plasticity in response to LDRT, suggesting that the nanomechanical properties of the tumor may be predictive of therapeutic response. Discussion: The findings of this study highlight the transformative potential of incorporating LDRT as a precursor to adoptive T-cell therapy in solid tumors. By promoting CAR-T and pmel+ T-cell infiltration into the tumor microenvironment, LDRT enhanced tumor control and improved survival outcomes, offering a promising strategy to overcome the challenges associated with CAR-T therapy in solid tumors. Additionally, the changes in tumor nanomechanics observed through AFM suggest that tumor stiffness and plasticity could be biomarkers for predicting treatment outcomes. These results support further investigation into the clinical application of this combined approach to improve the efficacy of cellbased therapies in patients with solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. 216 Tissue nanomechanical signature as a novel, clinically translatable predictor of response to immunotherapy combined with low-dose radiation
- Author
-
Nizzero, Sara, primary, Srivastava, Gitika, additional, Puebla-Osorio, Nahum, additional, Wasley, Mark, additional, Mitchell, Joylise, additional, Huang, Ailing, additional, Riad, Thomas, additional, Ndiaye, Papa Diogop, additional, Gachechiladze, Mariam, additional, Oertle, Philipp, additional, Appenzeller, Tobias, additional, Cristini, Vittorio, additional, Loparic, Marko, additional, Plodinec, Marija, additional, and Welsh, James, additional
- Published
- 2023
- Full Text
- View/download PDF
4. Combining a nanoparticle-mediated immunoradiotherapy with dual blockade of LAG3 and TIGIT improves the treatment efficacy in anti-PD1 resistant lung cancer
- Author
-
Sezen, Duygu (ORCID 0000-0002-4505-2280 & YÖK ID 170535), Hu, Yun; Paris, Sebastien; Bertolet, Genevieve; Barsoumian, Hampartsoum B.; He, Kewen; Chen, Dawei; Wasley, Mark; Da Silva, Jordan; Mitchell, Joylise A.; Voss, Tiffany A.; Masrorpour, Fatemeh; Leyton, Claudia Kettlun; Yang, Liangpeng; Leuschner, Carola; Puebla-Osorio, Nahum; Gandhi, Saumil; Quynh-Nhu Nguyen; Cortez, Maria Angelica; Welsh, James W., School of Medicine, Sezen, Duygu (ORCID 0000-0002-4505-2280 & YÖK ID 170535), Hu, Yun; Paris, Sebastien; Bertolet, Genevieve; Barsoumian, Hampartsoum B.; He, Kewen; Chen, Dawei; Wasley, Mark; Da Silva, Jordan; Mitchell, Joylise A.; Voss, Tiffany A.; Masrorpour, Fatemeh; Leyton, Claudia Kettlun; Yang, Liangpeng; Leuschner, Carola; Puebla-Osorio, Nahum; Gandhi, Saumil; Quynh-Nhu Nguyen; Cortez, Maria Angelica; Welsh, James W., and School of Medicine
- Abstract
Background: while improvements in immunoradiotherapy have significantly improved outcomes for cancer patients, this treatment approach has nevertheless proven ineffective at controlling the majority of malignancies. One of the mechanisms of resistance to immunoradiotherapy is that immune cells may be suppressed via the myriad of different immune checkpoint receptors. Therefore, simultaneous blockade of multiple immune checkpoint receptors may enhance the treatment efficacy of immunoradiotherapy. Methods: we combined NBTXR3-enhanced localized radiation with the simultaneous blockade of three different checkpoint receptors: PD1, LAG3, and TIGIT, and tested the treatment efficacy in an anti-PD1-resistant lung cancer model in mice. 129 Sv/Ev mice were inoculated with fifty thousand alpha PD1-resistant 344SQR cells in the right leg on day 0 to establish primary tumors and with the same number of cells in the left leg on day 4 to establish the secondary tumors. NBTXR3 was intratumorally injected into the primary tumors on day 7, which were irradiated with 12 Gy on days 8, 9, and 10. Anti-PD1 (200 mu g), alpha LAG3 (200 mu g), and alpha TIGIT (200 mu g) were given to mice by intraperitoneal injections on days 5, 8, 11, 14, 21, 28, 35, and 42. Results: this nanoparticle-mediated combination therapy is effective at controlling the growth of irradiated and distant unirradiated tumors, enhancing animal survival, and is the only one that led to the destruction of both tumors in approximately 30% of the treated mice. Corresponding with this improved response is robust activation of the immune response, as manifested by increased numbers of immune cells along with a transcriptional signature of both innate and adaptive immunity within the tumor. Furthermore, mice treated with this combinatorial therapy display immunological memory response when rechallenged by the same cancer cells, preventing tumor engraftment. Conclusion: our results strongly attest to the efficacy and validity, This work was supported by Cancer Center Support (Core) Grant CA016672 to The University of Texas MD Anderson Cancer Center; the Goodwin family research fund; the family of M. Adnan Hamed, and the Orr Family Foundation to MD Anderson Cancer Center's Thoracic Radiation Oncology program; an MD Anderson Knowledge Gap award; Nanobiotix.
- Published
- 2022
5. Combining a nanoparticle-mediated radioimmunotherapy with dual blockade of LAG3 and TIGIT improves the treatment efficacy in anti-PD1 resistant lung cancer
- Author
-
Hu, Yun, primary, Paris, Sébastien, additional, Bertolet, Genevieve, additional, Barsoumian, Hampartsoum, additional, He, Kewen, additional, Sezen, Duygu, additional, Chen, Dawei, additional, Wasley, Mark, additional, SILVA, Jordan DA, additional, Mitchell, Joylise A, additional, Voss, Tiffany A, additional, Masrorpour, Fatemeh, additional, Leyton, Claudia Kettlun, additional, Yang, Liangpeng, additional, Leuschner, Carola, additional, Puebla-Osorio, Nahum, additional, Gandhi, Saumil, additional, Nguyen, Quynh-Nhu, additional, Cortez, Maria Angelica, additional, and Welsh, James W., additional
- Published
- 2022
- Full Text
- View/download PDF
6. Additional file 2 of Combining a nanoparticle-mediated immunoradiotherapy with dual blockade of LAG3 and TIGIT improves the treatment efficacy in anti-PD1 resistant lung cancer
- Author
-
Hu, Yun, Paris, Sébastien, Bertolet, Genevieve, Barsoumian, Hampartsoum B., He, Kewen, Sezen, Duygu, Chen, Dawei, Wasley, Mark, SILVA, Jordan DA, Mitchell, Joylise A., Voss, Tiffany A., Masrorpour, Fatemeh, Leyton, Claudia Kettlun, Yang, Liangpeng, Leuschner, Carola, Puebla-Osorio, Nahum, Gandhi, Saumil, Nguyen, Quynh-Nhu, Cortez, Maria Angelica, and Welsh, James W.
- Abstract
Additional file 2: Table S1. Log2 fold change and the function of the genes which were significantly up-regulated in the unirradiated tumors treated with NBTXR3+XRT+PLT vs. NBTXR3+XRT+αPD1.
- Published
- 2022
- Full Text
- View/download PDF
7. Additional file 1 of Combining a nanoparticle-mediated immunoradiotherapy with dual blockade of LAG3 and TIGIT improves the treatment efficacy in anti-PD1 resistant lung cancer
- Author
-
Hu, Yun, Paris, Sébastien, Bertolet, Genevieve, Barsoumian, Hampartsoum B., He, Kewen, Sezen, Duygu, Chen, Dawei, Wasley, Mark, SILVA, Jordan DA, Mitchell, Joylise A., Voss, Tiffany A., Masrorpour, Fatemeh, Leyton, Claudia Kettlun, Yang, Liangpeng, Leuschner, Carola, Puebla-Osorio, Nahum, Gandhi, Saumil, Nguyen, Quynh-Nhu, Cortez, Maria Angelica, and Welsh, James W.
- Abstract
Additional file 1: Figure S1. Log2 fold change in the expression of TIGIT and LAG3 in tumors treated with NBTXR3 + XRT + αPD1. The 344SQR cells (5 × 104) were subcutaneously injected into the right legs of the 129/SvEv syngeneic female mice (n = 4, 8–12 weeks old) on day 0 to establish the “primary” tumor and into the left legs on day 4 to establish the “secondary” tumor. The primary tumors were intratumorally injected with NBTXR3 on day 7, followed by three fractions of 12 Gy radiation on days 8, 9, and 10. αPD1 (200 µg) was intraperitoneally injected into the mice on days 5, 8, 11, and 14. The expression of LAG3 and TIGIT in the irradiated and unirradiated tumors harvested on day 21 were measured by Nanostring. The mice inoculated with both primary and secondary tumors but did not receive any treatment served as control. P < 0.05 was considered statistically significant. **P < 0.01, ***P < 0.001. Figure S2. Individual tumor growth curves in mice receiving treatment as indicated in Fig. 1.Figure S3. Blockade of LAG3 and TIGIT with the combination of NBTXR3 + XRT + αPD1 reduces the number of lung metastases. The mice (n = 5) were treated with various combination therapies, as illustrated in Fig. 1, and were euthanized on day 21. The lungs were harvested and stored in Bouin’s fixative solution for three days, after which metastatic nodules were counted. The number of lung metastases was compared by t-test and were expressed as mean ± standard error of the mean (SEM). P < 0.05 was considered statistically significant. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, NS, not significant. Figure S4. Body weight of mice (n = 5) treated with combination therapy of NBTXR3 + XRT + αPD1 + αLAG3 + αTIGIT as shown ing Fig. 1A. Figure S5. Dual blockade of TIGIT and LAG3 promotes proliferation of CD4+ and CD8+ T cells. (A) Representative flow cytometry images of Ki67+CD4+ and Ki67+CD8+ T in primary tumors. (B) Representative flow cytometry images of Ki67+CD4+ and Ki67+CD8+ T cells in secondary tumors. (C) Representative flow cytometry images of Ki67+CD4+ and Ki67+CD8+ T cells in the blood. The mice (n = 5) were treated with various combination therapies, including XRT + αPD1, NBTXR3 + XRT + αPD1, NBTXR3 + XRT + αPD1 + αLAG3, NBTXR3 + XRT + αPD1 + αTIGIT, and NBTXR3 + XRT + PLT as indicated in Fig. 1A and were sacrificed on day 21. The mice which were inoculated with tumors only served as control. Immune cells from primary tumors, secondary tumors, and blood were processed and stained with αCD45-APC-Cy7, αCD3-PE-Cy7, αCD4-alexa 700, αCD8-PercpCy5.5, and αKi67-alexa 647. The cells were run with a Gallios Flow Cytometer (Beckman Coulter) and analyzed with Kaluza software Version 2.1. Figure S6. Flow cytometry gating strategy in Fig. 3. Figure S7. Nanostring cell score of various immune cells treated with different combinations of NBTXR3, XRT, αPD1, αLAG3 and αTIGIT in both primary and secondary tumors. Mice (n = 4) were treated with various combination therapies as described in Fig. 1 and were euthanized 11 d post last fraction of radiation. The RNA from the irradiated and unirradiated tumors was extracted, and the immune-related genes were measured with a nCounter PanCancer Immune Profiling Panel and a nCounter MAX Analysis System. The data were analyzed with the PanCancer Immune Profiling Advanced Analysis Module. The cell scores were compared by t-test and were expressed as mean ± standard error of the mean (SEM). P < 0.05 was considered statistically significant. ***P < 0.001, NS, not significant. Figure S8. Multiple immune-related genes are significantly differentially expressed in the tumors of mice following supplementation of NBTXR3 + XRT + αPD1 with αLAG3, αTIGIT, or both. Statistical significance was assessed via a two-tailed t-test, with a p < 0.05. A Heatmap of genes significantly up- or down-regulated in at least one of the three treatment groups relative to NBTXR3 + XRT + αPD-1 in the primary tumor. B Heatmap of genes significantly up- or down-regulated in at least one of the three treatment groups relative to NBTXR3 + XRT + αPD1 in the secondary tumor. Two hundred and forty-six (246) such genes were identified and are here ranked alphabetically. C Genes identified as significantly upregulated in NBTXR3 + XRT + PLT-treated mice compared to NBTXR3 + XRT + αPD1-treated mice were manually grouped according to their established function. Functional groups were plotted according to the log2 fold change from baseline (NBTXR3 + XRT + αPD1) of their constituent genes, represented in aggregate via violin plot.
- Published
- 2022
- Full Text
- View/download PDF
8. 575 Dual blockade of LAG3 and TIGIT improves the treatment efficacy of a nanoparticle-mediated immunoradiation in anti-PD1 resistant lung cancer in mice
- Author
-
Hu, Yun, primary, Welsh, James, additional, Paris, Sebastien, additional, Bertolet, Genevieve, additional, Barsoumian, Hampartsoum, additional, Schuda, Lily, additional, He, Kewen, additional, Sezen, Duygu, additional, Wasley, Mark, additional, Mitchell, Joylise, additional, Voss, Tiffany, additional, Masrorpour, Fatemeh, additional, Jordan, SILVA, additional, Leyton, Claudia Kettlun, additional, Yang, Liangpeng, additional, Puebla-Osorio, Nahum, additional, Gandhi, Saumil, additional, Nguyen, Quynh-Nhu, additional, and Cortez, Angelica, additional
- Published
- 2021
- Full Text
- View/download PDF
9. Combining a nanoparticle-mediated immunoradiotherapy with dual blockade of LAG3 and TIGIT improves the treatment efficacy in anti-PD1 resistant lung cancer
- Author
-
Yun, Hu, Sébastien, Paris, Genevieve, Bertolet, Hampartsoum B, Barsoumian, Kewen, He, Duygu, Sezen, Dawei, Chen, Mark, Wasley, Jordan DA, Silva, Joylise A, Mitchell, Tiffany A, Voss, Fatemeh, Masrorpour, Claudia Kettlun, Leyton, Liangpeng, Yang, Carola, Leuschner, Nahum, Puebla-Osorio, Saumil, Gandhi, Quynh-Nhu, Nguyen, Maria Angelica, Cortez, James W, Welsh, Sezen, Duygu (ORCID 0000-0002-4505-2280 & YÖK ID 170535), Hu, Yun, Paris, Sebastien, Bertolet, Genevieve, Barsoumian, Hampartsoum B., He, Kewen, Chen, Dawei, Wasley, Mark, Da Silva, Jordan, Mitchell, Joylise A., Voss, Tiffany A., Masrorpour, Fatemeh, Leyton, Claudia Kettlun, Yang, Liangpeng, Leuschner, Carola, Puebla-Osorio, Nahum, Gandhi, Saumil, Quynh-Nhu Nguyen, Cortez, Maria Angelica, Welsh, James W., and School of Medicine
- Subjects
Lung Neoplasms ,Biomedical Engineering ,Pharmaceutical Science ,Medicine (miscellaneous) ,Antineoplastic Agents ,Bioengineering ,Radioimmunotherapy ,Lymphocyte Activation Gene 3 Protein ,Applied Microbiology and Biotechnology ,Mice ,Treatment Outcome ,Nanoparticle ,NBTXR3 ,Immunoradiotherapy ,Radiotherapy ,Checkpoint blockade ,Anti-PD1 resistance ,Antigens, CD ,Biotechnology and applied microbiology ,Nanoscience and nanotechnology ,Animals ,Humans ,Nanoparticles ,Molecular Medicine ,Receptors, Immunologic - Abstract
Background: while improvements in immunoradiotherapy have significantly improved outcomes for cancer patients, this treatment approach has nevertheless proven ineffective at controlling the majority of malignancies. One of the mechanisms of resistance to immunoradiotherapy is that immune cells may be suppressed via the myriad of different immune checkpoint receptors. Therefore, simultaneous blockade of multiple immune checkpoint receptors may enhance the treatment efficacy of immunoradiotherapy. Methods: we combined NBTXR3-enhanced localized radiation with the simultaneous blockade of three different checkpoint receptors: PD1, LAG3, and TIGIT, and tested the treatment efficacy in an anti-PD1-resistant lung cancer model in mice. 129 Sv/Ev mice were inoculated with fifty thousand alpha PD1-resistant 344SQR cells in the right leg on day 0 to establish primary tumors and with the same number of cells in the left leg on day 4 to establish the secondary tumors. NBTXR3 was intratumorally injected into the primary tumors on day 7, which were irradiated with 12 Gy on days 8, 9, and 10. Anti-PD1 (200 mu g), alpha LAG3 (200 mu g), and alpha TIGIT (200 mu g) were given to mice by intraperitoneal injections on days 5, 8, 11, 14, 21, 28, 35, and 42. Results: this nanoparticle-mediated combination therapy is effective at controlling the growth of irradiated and distant unirradiated tumors, enhancing animal survival, and is the only one that led to the destruction of both tumors in approximately 30% of the treated mice. Corresponding with this improved response is robust activation of the immune response, as manifested by increased numbers of immune cells along with a transcriptional signature of both innate and adaptive immunity within the tumor. Furthermore, mice treated with this combinatorial therapy display immunological memory response when rechallenged by the same cancer cells, preventing tumor engraftment. Conclusion: our results strongly attest to the efficacy and validity of combining nanoparticle-enhanced radio-therapy and simultaneous blockade of multiple immune checkpoint receptors and provide a pre-clinical rationale for investigating its translation into human patients., This work was supported by Cancer Center Support (Core) Grant CA016672 to The University of Texas MD Anderson Cancer Center; the Goodwin family research fund; the family of M. Adnan Hamed, and the Orr Family Foundation to MD Anderson Cancer Center's Thoracic Radiation Oncology program; an MD Anderson Knowledge Gap award; Nanobiotix.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.