128 results on '"Intratumoral Therapy"'
Search Results
2. Current Role and Status for Intratumoral Injection Therapies in Metastatic Melanoma.
- Author
-
Haugh, Alexandra M. and Daud, Adil I.
- Published
- 2024
- Full Text
- View/download PDF
3. Initial Safety and Feasibility Results From a Phase 1, Diagnose-and-Treat Trial of Neoadjuvant Intratumoral Cisplatin for Stage IV NSCLC
- Author
-
Farrah B. Khan, MD, Pamela C. Gibson, MD, Scott Anderson, MD, Sarah Wagner, BS, Bernard F. Cole, PhD, Peter Kaufman, MD, and C. Matthew Kinsey, MD, MPH
- Subjects
Intratumoral therapy ,Cisplatin ,Endobronchial ultrasound-guided transbronchial needle injection ,Lung cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Neoadjuvant intratumoral cisplatin has the potential to generate substantial cytotoxicity and immune priming within the tumor environment, while minimizing systemic, off-target, adverse events. We initiated a phase 1A, 3+3 dose-ranging study of neoadjuvant, intratumoral cisplatin, delivered through endobronchial ultrasound bronchoscopy, in the same procedure as the initial diagnosis. There were no dose-limiting toxicity identified at the 20mg level
- Published
- 2024
- Full Text
- View/download PDF
4. Engaging Pattern Recognition Receptors in Solid Tumors to Generate Systemic Antitumor Immunity
- Author
-
Brown, Michael, Rosen, Steven T., Series Editor, and Hays, Priya, editor
- Published
- 2022
- Full Text
- View/download PDF
5. An open-label, single-arm pilot study of EUS-guided brachytherapy with phosphorus-32 microparticles in combination with gemcitabine +/- nab-paclitaxel in unresectable locally advanced pancreatic cancer (OncoPaC-1): Technical details and study protocol
- Author
-
Bhutani, Manoop S, Klapman, Jason B, Tuli, Richard, El-Haddad, Ghassan, Hoffe, Sarah, Wong, Franklin CL, Chasen, Beth, Fogelman, David R, Lo, Simon K, Nissen, Nicholas N, Hendifar, Andrew E, Varadhachary, Gauri, Katz, Matthew HG, Erwin, William D, Koay, Eugene J, Tamm, Eric P, Singh, Ben S, Mehta, Rutika, Wolff, Robert A, Soman, Ashish, Cazacu, Irina M, and Herman, Joseph M
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Pancreatic Cancer ,Digestive Diseases ,Clinical Research ,Orphan Drug ,Rare Diseases ,Cancer ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Brachytherapy ,EUS ,chemotherapy ,endoscopy ,fine needle injection ,intratumoral therapy ,neoplasia ,pancreatic cancer ,phosphorus-32 ,radiotherapy - Abstract
Current treatment options for patients with unresectable locally advanced pancreatic cancer (LAPC) include chemotherapy alone or followed by chemoradiation or stereotactic body radiotherapy. However, the prognosis for these patients remains poor, with a median overall survival
- Published
- 2020
6. Advances in Intralesional Therapy for Locoregionally Advanced and Metastatic Melanoma: Five Years of Progress.
- Author
-
DePalo, Danielle K. and Zager, Jonathan S.
- Subjects
- *
MELANOMA treatment , *CYTOKINES , *DRUG efficacy , *INJECTIONS , *IMMUNE checkpoint inhibitors , *MANUSCRIPTS , *COMBINATION drug therapy , *METASTASIS , *TREATMENT effectiveness , *BIOTHERAPY , *IMMUNOTHERAPY , *TOLL-like receptors - Abstract
Simple Summary: The prognosis for patients with locoregionally advanced and metastatic melanoma remains poor despite advances in systemic therapy. By delivering therapeutics directly to the site(s) of disease, intralesional therapies have the advantage of delivering the oncolytic agent directly into the metastatic melanoma while minimizing systemic side effects and resistance. Within the past 5 years, numerous potential intratumoral therapies have been investigated, though few have reached phase 2 clinical trials. We present a discussion of the scientific rationale for and status of intralesional therapies that have reached phase 2 or later clinical trials within the past 5 years in order to inform providers about current and upcoming intralesional therapeutic options for advanced melanoma. Locoregionally advanced and metastatic melanoma are complex diagnoses with a variety of available treatment options. Intralesional therapy for melanoma has been under investigation for decades; however, it has advanced precipitously in recent years. In 2015, the Food and Drug Administration (FDA) approved talimogene laherparepvec (T-VEC), the only FDA-approved intralesional therapy for advanced melanoma. There has been significant progress since that time with other oncolytic viruses, toll-like receptor agonists, cytokines, xanthene dyes, and immune checkpoint inhibitors all under investigation as intralesional agents. Further to this, there has been exploration of numerous combinations of intralesional therapies and systemic therapies as various lines of therapy. Several of these combinations have been abandoned due to their lack of efficacy or safety concerns. This manuscript presents the various types of intralesional therapies that have reached phase 2 or later clinical trials in the past 5 years, including their mechanism of action, therapeutic combinations under investigation, and published results. The intention is to provide an overview of the progress that has been made, discuss ongoing trials worth following, and share our opinions on opportunities for further advancement. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Leading Edge: Intratumor Delivery of Monoclonal Antibodies for the Treatment of Solid Tumors.
- Author
-
Blanco, Ester, Chocarro, Luisa, Fernández-Rubio, Leticia, Bocanegra, Ana, Arasanz, Hugo, Echaide, Miriam, Garnica, Maider, Piñeiro-Hermida, Sergio, Kochan, Grazyna, and Escors, David
- Subjects
- *
MONOCLONAL antibodies , *TUMOR treatment , *IMMUNE checkpoint proteins , *GENE therapy , *TREATMENT effectiveness , *NANOMEDICINE - Abstract
Immunotherapies based on immune checkpoint blockade have shown remarkable clinical outcomes and durable responses in patients with many tumor types. Nevertheless, these therapies lack efficacy in most cancer patients, even causing severe adverse events in a small subset of patients, such as inflammatory disorders and hyper-progressive disease. To diminish the risk of developing serious toxicities, intratumor delivery of monoclonal antibodies could be a solution. Encouraging results have been shown in both preclinical and clinical studies. Thus, intratumor immunotherapy as a new strategy may retain efficacy while increasing safety. This approach is still an exploratory frontier in cancer research and opens up new possibilities for next-generation personalized medicine. Local intratumor delivery can be achieved through many means, but an attractive approach is the use of gene therapy vectors expressing mAbs inside the tumor mass. Here, we summarize basic, translational, and clinical results of intratumor mAb delivery, together with descriptions of non-viral and viral strategies for mAb delivery in preclinical and clinical development. Currently, this is an expanding research subject that will surely play a key role in the future of oncology. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Emerging immunotherapeutic strategies for the treatment of breast cancer.
- Author
-
Huppert, Laura A., Mariotti, Veronica, Chien, A. Jo, and Soliman, Hatem H.
- Abstract
Immunotherapy has resulted in unprecedented gains in long-term outcomes for many cancer types and has revolutionized the treatment landscape of solid tumor oncology. Checkpoint inhibition in combination with chemotherapy has proven to be effective for the treatment of a subset of advanced triple-negative breast cancer in the first-line setting. This initial success is likely just the tip of the iceberg as there is much that remains unknown about how to best harness the immune system as a therapeutic strategy in all breast cancer subtypes. Therefore, numerous ongoing studies are currently underway to evaluate the safety and efficacy of immunotherapy in breast cancer. In this review, we will discuss emerging immunotherapeutic strategies for breast cancer treatment including the following: (1) Intratumoral therapies, (2) Anti-tumor vaccines, (3) B-specific T-cell engagers, and (4) Chimeric antigen receptor T-cell therapy, and (5) Emerging systemic immunotherapy strategies. For each topic, we will review the existing preclinical and clinical literature, discuss ongoing clinical trials, and highlight future directions in the field. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. Neue Entwicklungen in der Immunonkologie.
- Author
-
Ochsenreither, S.
- Abstract
Copyright of Der Onkologe is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2021
- Full Text
- View/download PDF
10. Intratumoral therapies in head and neck squamous cell carcinoma: A systematic review and future perspectives.
- Author
-
Jiménez-Labaig, Pablo, Rullan, Antonio, Braña, Irene, Hernando-Calvo, Alberto, Moreno, Victor, Doger, Bernard, Bitar, George, Ap Dafydd, Derfel, Melcher, Alan, and Harrington, Kevin J.
- Abstract
• Head and neck squamous cell carcinoma presents an ideal scenario for intratumoral therapies, due to its local recurrence pattern and frequent superficial extension. • Its landscape is dominated by phase I-II studies, including chemotherapies, oncolytic viruses, bacteria, plasmids, immunotherapies, EGFR-targeted therapies and radioenhancer particles. • Some antineoplastic agents, when directly injected, offer the potential for tumor shrinkage while reducing systemically exposure. • Necrosis, fistula formation, post-injection dysphagia and bleedings are the major concerns. • Further research is needed to improve patient selection, ensure safety in injections, and conduct randomized controlled trials. Head and neck squamous cell carcinoma (HNSCC) presents an ideal scenario for intratumoral therapies (IT), due to its local recurrence pattern and frequent superficial extension. IT therapies aim to effect tumor regression by directly injecting antineoplastic agents into lesions. However, there is a lack of updated evidence regarding IT therapies in HNSCC. A systematic literature search (CRD42023462291) was conducted using WebOfScience, ClinicalTrials.gov , and conference abstracts from ESMO and ASCO, identifying for IT clinical trials in patients with HNSCC, from database creation to September 12th, 2023. Efficacy as well as safety (grade ≥ 3 treatment-related adverse events[trAEs]) were reported. After evaluation of 1180 articles identified by the systematic search, 31 studies treating 948 patients were included. IT injectables were categorized as chemotherapies with or without electroporation (k = 4, N = 268), oncolytic viruses, plasmids, and bacteria-based (k = 16, N = 446), immunotherapies and EGFR-based therapies (k = 5, N = 160), radioenhancer particles (k = 2, N = 68), and calcium electroporation (k = 1, n = 6). EGFR-antisense plasmids, NBTXR3 radioenhancer and immune innate agonists show best overall response rates, at 83 %, 81 % and 44 % respectively. Eleven (35 %) studies added systemic therapy or radiotherapy to the IT injections. No study used predictive biomarkers to guide patient selection. 97 % studies were phase I-II. Safety-wise, electroporation and epinephrine-based injectable trials had significant local symptoms such as necrosis, fistula formation and post-injection dysphagia. Treatment-related tumor haemorrhages of various grades were described in several trials. Grade ≥ 3 trAEs attributable to the other therapies mainly comprised general symptoms such as fatigue. There were 3 injectable-related deaths across the systematic review. This is the first review to summarize all available evidence of IT in HNSCC. As of today, IT therapies lack sufficient evidence to recommend their use in clinical practice. Continuing research on potential molecules, patient selection, safe administration of injections and controlled randomized trials are needed to assess their added benefit. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Synthetic RIG-I agonist-mediated cancer immunotherapy synergizes with MAP kinase inhibition against BRAF-mutated melanoma.
- Author
-
Grützner C, Tan YP, Müller P, Schlee-Guimaraes TM, Jentzsch M, Schmid-Burgk JL, Renn M, Behrendt R, and Hartmann G
- Abstract
The implementation of targeted molecular therapies and immunotherapy in melanoma vastly improved the therapeutic outcome in patients with limited efficacy of surgical intervention. Nevertheless, a large fraction of patients with melanoma still remain refractory or acquire resistance to these new forms of treatment, illustrating a need for improvement. Here, we report that the clinically relevant combination of mitogen-activated protein (MAP) kinase pathway inhibitors dabrafenib and trametinib synergize with RIG-I agonist-induced immunotherapy to kill BRAF-mutated human and mouse melanoma cells. Kinase inhibition did not compromise the agonist-induced innate immune response of the RIG-I pathway in host immune cells. In a melanoma transplantation mouse model, the triple therapy outperformed individual therapies. Our study suggests that agonist-induced activation of RIG-I with its synthetic ligand 3pRNA could vastly improve tumor control in a substantial fraction of patients with melanoma receiving MAP kinase inhibitors., Competing Interests: G.H. is an inventor on a patent covering synthetic RIG-I ligands. M.R. and G.H. were co-founders of Rigontec GmbH. J.L.S.-B. is a co-founder and shareholder of LAMPseq Diagnostics., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
12. An experimental model of anti-PD-1 resistance exhibits activation of TGFß and Notch pathways and is sensitive to local mRNA immunotherapy
- Author
-
Marie Bernardo, Tatiana Tolstykh, Yu-an Zhang, Dinesh S. Bangari, Hui Cao, Kerstin A. Heyl, Joon Sang Lee, Natalia V. Malkova, Katie Malley, Eladio Marquez, Jack Pollard, Hui Qu, Errin Roberts, Sue Ryan, Kuldeep Singh, Fangxian Sun, Emma Wang, Keith Bahjat, Dmitri Wiederschain, and Timothy R. Wagenaar
- Subjects
checkpoint blockade resistance ,syngeneic tumor model ,cancer immunotherapy ,intratumoral therapy ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Immune checkpoint blockade elicits durable anti-cancer responses in the clinic, however a large proportion of patients do not benefit from treatment. Several mechanisms of innate and acquired resistance to checkpoint blockade have been defined and include mutations of MHC I and IFNγ signaling pathways. However, such mutations occur in a low frequency of patients and additional mechanisms have yet to be elucidated. In an effort to better understand acquired resistance to checkpoint blockade, we generated a mouse tumor model exhibiting in vivo resistance to anti-PD-1 antibody treatment. MC38 tumors acquired resistance to PD-1 blockade following serial in vivo passaging. Lack of sensitivity to PD-1 blockade was not attributed to dysregulation of PD-L1 or β2M expression, as both were expressed at similar levels in parental and resistant cells. Similarly, IFNγ signaling and antigen processing and presentation pathways were functional in both parental and resistant cell lines. Unbiased gene expression analysis was used to further characterize potential resistance mechanisms. RNA-sequencing revealed substantial differences in global gene expression, with tumors resistant to anti-PD-1 displaying a marked reduction in expression of immune-related genes relative to parental MC38 tumors. Indeed, resistant tumors exhibited reduced immune infiltration across multiple cell types, including T and NK cells. Pathway analysis revealed activation of TGFβ and Notch signaling in anti-PD-1 resistant tumors, and activation of these pathways was associated with poorer survival in human cancer patients. While pharmacological inhibition of TGFβ and Notch in combination with PD-1 blockade decelerated tumor growth, a local mRNA-based immunotherapy potently induced regression of resistant tumors, resulting in complete tumor remission, and resensitized tumors to treatment with anti-PD-1. Overall, this study describes a novel anti-PD-1 resistant mouse tumor model and underscores the role of two well-defined signaling pathways in response to immune checkpoint blockade. Furthermore, our data highlights the potential of intratumoral mRNA therapy in overcoming acquired resistance to PD-1 blockade.
- Published
- 2021
- Full Text
- View/download PDF
13. An experimental model of anti-PD-1 resistance exhibits activation of TGFβ and Notch pathways and is sensitive to local mRNA immunotherapy.
- Author
-
Bernardo, Marie, Tolstykh, Tatiana, Yu-an Zhang, Bangari, Dinesh S., Hui Cao, Heyl, Kerstin A., Joon Sang Lee, Malkova, Natalia V., Malley, Katie, Marquez, Eladio, Pollard, Jack, Hui Qu, Roberts, Errin, Ryan, Sue, Singh, Kuldeep, Sun, Fangxian, Wang, Emma, Bahjat, Keith, Wiederschain, Dmitri, and Wagenaar, Timothy R.
- Subjects
IMMUNE checkpoint proteins ,KILLER cells ,NATURAL immunity ,MESSENGER RNA ,ANTIGEN processing - Abstract
Immune checkpoint blockade elicits durable anti-cancer responses in the clinic, however a large proportion of patients do not benefit from treatment. Several mechanisms of innate and acquired resistance to checkpoint blockade have been defined and include mutations of MHC I and IFNγ signaling pathways. However, such mutations occur in a low frequency of patients and additional mechanisms have yet to be elucidated. In an effort to better understand acquired resistance to checkpoint blockade, we generated a mouse tumor model exhibiting in vivo resistance to anti-PD-1 antibody treatment. MC38 tumors acquired resistance to PD-1 blockade following serial in vivo passaging. Lack of sensitivity to PD-1 blockade was not attributed to dysregulation of PD-L1 or β2M expression, as both were expressed at similar levels in parental and resistant cells. Similarly, IFNγ signaling and antigen processing and presentation pathways were functional in both parental and resistant cell lines. Unbiased gene expression analysis was used to further characterize potential resistance mechanisms. RNA-sequencing revealed substantial differences in global gene expression, with tumors resistant to anti-PD-1 displaying a marked reduction in expression of immune-related genes relative to parental MC38 tumors. Indeed, resistant tumors exhibited reduced immune infiltration across multiple cell types, including T and NK cells. Pathway analysis revealed activation of TGFβ and Notch signaling in anti-PD-1 resistant tumors, and activation of these pathways was associated with poorer survival in human cancer patients. While pharmacological inhibition of TGFβ and Notch in combination with PD-1 blockade decelerated tumor growth, a local mRNA-based immunotherapy potently induced regression of resistant tumors, resulting in complete tumor remission, and resensitized tumors to treatment with anti-PD-1. Overall, this study describes a novel anti-PD-1 resistant mouse tumor model and underscores the role of two well-defined signaling pathways in response to immune checkpoint blockade. Furthermore, our data highlights the potential of intratumoral mRNA therapy in overcoming acquired resistance to PD-1 blockade. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
14. Intratumoral infection by CMV may change the tumor environment by directly interacting with tumor-associated macrophages to promote cancer immunity
- Author
-
Dan A. Erkes, Nicole A. Wilski, and Christopher M. Snyder
- Subjects
cytomegalovirus ,intratumoral therapy ,tumor associated macrophages ,Immunologic diseases. Allergy ,RC581-607 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Cytomegalovirus (CMV) is a herpesvirus that induces an extremely robust and sustained immune response. For this reason, CMV has been proposed as a vaccine vector to promote immunity to both pathogens and cancer. However, exploration of CMV as a vaccine vector is at an early stage and there are many questions. Using a mouse melanoma model, we recently found that a CMV-based vaccine induced large populations of melanoma-specific T cells, but was not effective at slowing tumor growth unless it was injected directly into the tumor. These surprising results have led us to hypothesize that CMV may be adept at modulating the tumor micro-environment through its infection of macrophages. Importantly, injection of CMV into the growing tumor synergized with blockade of the PD-1 checkpoint to clear well-established tumors. Here, we discuss our results in the context of CMV-based vaccines for pathogens and cancer.
- Published
- 2017
- Full Text
- View/download PDF
15. Novel Cytokines for Immunotherapy of Melanoma
- Author
-
Bhatia, Shailender, Thompson, John A., Gajewski, Thomas F., editor, and Hodi, F. Stephen, editor
- Published
- 2012
- Full Text
- View/download PDF
16. Real-World Use of Talimogene Laherparepvec in German Patients with Stage IIIB to IVM1a Melanoma: A Retrospective Chart Review and Physician Survey.
- Author
-
Mohr, Peter, Haferkamp, Sebastian, Pinter, Andreas, Weishaupt, Carsten, Huber, Margit A., Downey, Gerald, Öhrling, Katarina, Loquai, Carmen, and Louie, Karly S.
- Subjects
BIOTHERAPY ,COMPARATIVE studies ,HERPESVIRUSES ,IMMUNOTHERAPY ,INJECTIONS ,LONGITUDINAL method ,RESEARCH methodology ,MEDICAL cooperation ,MELANOMA ,RESEARCH ,EVALUATION research ,TREATMENT effectiveness ,RETROSPECTIVE studies - Abstract
Introduction: Talimogene laherparepvec is a first-in-class oncolytic immunotherapy for intratumoral injection with proven efficacy and tolerability in patients with unresectable early metastatic melanoma (stage IIIB-IVM1a) in the pivotal phase III OPTiM study. The objective was to characterize melanoma patients treated with talimogene laherparepvec in routine clinical practice in Germany.Methods: A retrospective chart review was conducted in unresectable stage IIIB-IVM1a melanoma patients. Data on demographics, disease and medical history, and use of talimogene laherparepvec were collected. A survey was also conducted to understand physician treatment decisions.Results: Data for 27 patients who initiated talimogene laherparepvec between June 2016 and July 2017 were analyzed (median age 68; stage IIIB/C disease 56%). All patients had prior surgery, and over half had repeated resections for recurrent disease (median 3). Overall, 48% of patients received at least one prior local treatment, mainly radiation therapy or electrochemotherapy. Talimogene laherparepvec was first-line systemic therapy in 63% of patients. The most frequent prior systemic treatment was immunotherapy (7/27 patients). At end of follow-up, 13 patients were still on talimogene laherparepvec and 14 patients had discontinued treatment. Among those who discontinued, 8 (57%) did not receive subsequent systemic therapy. Only one patient receiving first-line talimogene laherparepvec received a subsequent systemic therapy. Three patients stopped treatment because of no remaining injectable lesions. Median treatment duration was 22.1 weeks overall and 27.9 weeks in stage IIIB/C disease patients. Nearly all cutaneous lesions (93%) were injected with talimogene laherparepvec compared to subcutaneous (83%) and nodal lesions (77%). No new safety signals were reported. The main reasons given in the physician survey for treating with talimogene laherparepvec were good tolerability, overall efficacy, and lack of contraindications.Conclusion: Talimogene laherparepvec is now included as a routine treatment option for unresectable early metastatic melanoma in Germany. This study characterizes the first patients treated with talimogene laherparepvec in Europe and confirms the good tolerability observed in clinical trials.Trial Registration: EUPAS registry, EUPAS17410.Funding: Amgen Inc. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
17. Development of an adaptive electroporation system for intratumoral plasmid DNA delivery.
- Author
-
Brown, Douglas W., Bahrami, Arya J., Canton, David A., Mukhopadhyay, Anandaroop, Campbell, Jean S., Pierce, Robert H., and Connolly, Richard J.
- Subjects
- *
ELECTROPORATION , *PLASMIDS , *GENETIC code , *GENE delivery techniques , *INTERLEUKIN-12 - Abstract
Intratumoral electroporation of plasmid DNA encoding the proinflammatory cytokine interleukin 12 promotes innate and adaptive immune responses correlating with anti-tumor effects. Clinical electroporation conditions are fixed parameters optimized in preclinical tumors, which consist of cells implanted into skin. These conditions have little translatability to clinically relevant tumors, as implanted models cannot capture the heterogeneity encountered in genetically engineered mouse models or clinical tumors. Variables affecting treatment outcome include tumor size, degree of vascularization, fibrosis, and necrosis, which can result in suboptimal gene transfer and variable therapeutic outcomes. To address this, a feedback controlled electroporation generator was developed, which is capable of assessing the electrochemical properties of tissue in real time. Determination of these properties is accomplished by impedance spectroscopy and equivalent circuit model parameter estimation. Model parameters that estimate electrical properties of cell membranes are used to adjust electroporation parameters for each applied pulse. Studies performed in syngeneic colon carcinoma tumors (MC38) and spontaneous mammary tumors (MMTV-PyVT) demonstrated feedback-based electroporation is capable of achieving maximum expression of reporter genes with significantly less variability and applied energy. These findings represent an advancement to the practice of gene electro-transfer, as reducing variability and retaining transfected cell viability is paramount to treatment success. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
18. Endobronchial ultrasound-guided transbronchial needle injection of cisplatin results in dynamic changes in the tumor immune microenvironment.
- Author
-
DuComb EA, Collins CC, Cupak D, Wagner S, Khan FB, Budd RC, and Kinsey CM
- Subjects
- Humans, Cisplatin therapeutic use, Tumor Microenvironment, Endoscopic Ultrasound-Guided Fine Needle Aspiration methods, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms pathology
- Abstract
Rationale: Direct intratumoral delivery of cisplatin via endobronchial ultrasound guided-transbronchial needle injections (EBUS-TBNI) is a novel approach for salvage treatment of advanced stage non-small cell lung cancer (NSCLC). The goal of this study was to evaluate changes in the tumor immune microenvironment during the course of EBUS-TBNI cisplatin therapy., Methods: Under an IRB approved protocol, patients with recurrence after radiation therapy who were not receiving other cytotoxic therapy, were prospectively enrolled, and underwent weekly treatments with EBUS-TBNI with additional biopsies obtained for research. Needle aspiration was performed prior to cisplatin delivery at each procedure. Samples were evaluated by flow cytometry for the presence of immune cell types., Results: Three of the six patients responded to the therapy based on RECIST criteria. Compared to the pre-treatment baseline, intratumoral neutrophils increased in 5 of the 6 patients (p = 0.041), with an average increase of 27.1%, but was not associated with response. A lower pre-treatment CD8+/CD4+ ratio at baseline was associated with response (P = 0.01). Responders demonstrated a lower final proportion of PD-1+ CD8+ T cells compared to non-responders (8.6% vs. 62.3%, respectively, P<0.001. Lower doses of intratumoral cisplatin were associated with subsequent increases in CD8+ T cells within the tumor microenvironment (P = 0.008)., Conclusions: EBUS-TBNI cisplatin resulted in significant alterations in the tumor immune microenvironment. Further studies are needed to determine if the changes seen here generalize to larger cohorts., Competing Interests: Declaration of Competing Interest None., (Copyright © 2023 SPLF and Elsevier Masson SAS. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. Nanoformulation of CCL21 greatly increases its effectiveness as an immunotherapy for neuroblastoma
- Author
-
Don W. Coulter, Bailee H. Sliker, Benjamin T. Goetz, Madeline T. Olson, Kaitlin Smits, Alexandra E.J. Moffitt-Holida, Hamid Band, Brittney L. Dickey, Lynette M. Smith, Nuzhat Khan, Shelby M. Knoche, Svetlana Romanova, Brittany J. Poelaert, Aaron M. Mohs, Joyce C. Solheim, and Tatiana K. Bronich
- Subjects
endocrine system ,Chemokine ,Intratumoral Therapy ,medicine.medical_treatment ,Cell ,Pharmaceutical Science ,02 engineering and technology ,Ligands ,Article ,Mice ,Neuroblastoma ,03 medical and health sciences ,Immune system ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Effective treatment ,030304 developmental biology ,0303 health sciences ,Chemokine CCL21 ,biology ,business.industry ,Immunotherapy ,021001 nanoscience & nanotechnology ,medicine.disease ,medicine.anatomical_structure ,biology.protein ,Cancer research ,0210 nano-technology ,business ,CCL21 - Abstract
Neuroblastoma is the most commonly diagnosed extracranial solid tumor in children. The patients with aggressive metastatic disease or refractory/relapsed neuroblastoma currently face a dismally low chance of survival. Thus, there is a great need for more effective therapies for this illness. In previous studies, we, as well as others, showed that the immune cell chemoattractant C-C motif chemokine ligand 21 (CCL21) is effective as an intratumoral therapy able to slow the growth of cancers. In this current study, we developed and tested an injectable, slow-release, uniform, and optimally loaded alginate nanoformulation of CCL21 as a means to provide prolonged intratumoral treatment. The alginate-nanoformulated CCL21, when injected intratumorally into mice bearing neuroblastoma lesions, significantly prolonged survival and decreased the tumor growth rate compared to CCL21 alone, empty nanoparticles, or buffer. Notably, we also observed complete tumor clearance and subsequent full protection against tumor rechallenge in 33% of nanoformulated CCL21-treated mice. Greater intratumoral presence of nanoformulated CCL21, compared to free CCL21, at days 1 and 2 after treatment ended was confirmed through fluorescent labeling and tracking. Nanoformulated CCL21-treated tumors exhibited a general pattern of prolonged increases in anti-tumor cytokines and relatively lower levels of pro-tumor cytokines in comparison to tumors treated with CCL21 alone or buffer only. Thus, this novel nanoformulation of CCL21 is an effective treatment for neuroblastoma, and may have potential for the delivery of CCL21 to other types of solid tumors in the future and as a slow-release delivery modality for other immunotherapies.
- Published
- 2020
- Full Text
- View/download PDF
20. Bronchoscopic intratumoral injections of cisplatin and endostar as concomitants of standard chemotherapy to treat malignant central airway obstruction
- Author
-
Yue Li, Na Wang, Xuekun Wang, Xiaoping Yang, Bin Yin, Wenqing Jiang, and Xinai Yang
- Subjects
Male ,medicine.medical_specialty ,Lung Neoplasms ,Combination therapy ,Intratumoral Therapy ,medicine.medical_treatment ,Angiogenesis Inhibitors ,Antineoplastic Agents ,Injections, Intralesional ,03 medical and health sciences ,FEV1/FVC ratio ,0302 clinical medicine ,Bronchoscopy ,Carcinoma, Non-Small-Cell Lung ,Humans ,Medicine ,Aged ,030304 developmental biology ,Aged, 80 and over ,Cisplatin ,0303 health sciences ,Chemotherapy ,medicine.diagnostic_test ,business.industry ,General Medicine ,Middle Aged ,Debulking ,Recombinant Proteins ,Endostatins ,Airway Obstruction ,Treatment Outcome ,030220 oncology & carcinogenesis ,Quality of Life ,Female ,Radiology ,Airway ,business ,medicine.drug - Abstract
Study Purpose Malignant central airway obstruction (CAO) in non-small cell lung cancer (NSCLC) is associated with high morbidity and requires endobronchial palliative treatment to re-establish a free air passage. We investigate intratumoral therapy combining anti-angiogenic and cytotoxic as a feasible therapeutic modality to treat malignant CAO. Study Design Ten NSCLC subjects with symptomatic malignant CAO underwent endobronchial intratumoral cisplatin and Endostar co-injection after tumour debulking next to systemic cisplatin-based chemotherapy. Injection was performed immediately after debulking surgery and was then carried out on day 2, day 6 and day 10 past systemic chemotherapy. Nine subjects of control group constantly received traditional cisplatin-based chemotherapy. Bronchoscopy, CT scanning, histology, FEV1/FVC ratio, Karnofsky performance (KPS) and shortness of breath scores were analysed to assess therapeutic efficacy. Results All 10 subjects benefited from the intratumoral cisplatin and endostar co-injection and systemic chemotherapy combination therapy. Bronchoscopy and CT scanning analyses showed a massive airway widening after treatment. Increased KPS and reduced shortness of breath score were also observed. A substantial improvement of lung function was further confirmed by increased FEV1/FVC ratio. For subjects of control group, the improvement was moderate and obviously not as optimal as the 10 subjects with intratumoral injection. Conclusions We have shown that the intratumoral injection of cytotoxic cisplatin plus anti-angiogenic Endostar is an effective and safe adjuvant therapeutic option to treat malignant CAO in clinical practice. This time-staggered local and systemic treatment combination improves quality of life and clinical parameters, thus may provide a feasible therapeutic option for symptomatic CAO.
- Published
- 2020
- Full Text
- View/download PDF
21. Human intratumoral therapy: Linking drug properties and tumor transport of drugs in clinical trials
- Author
-
Chad Groer, Ruolin Lu, M. Laird Forrest, Aric Huang, Sebastian G. Huayamares, Melissa M. Pressnall, Brandon J. DeKosky, J. Daniel Griffin, and Cory Berkland
- Subjects
medicine.medical_treatment ,Intratumoral Therapy ,Pharmaceutical Science ,02 engineering and technology ,03 medical and health sciences ,Immune system ,Neoplasms ,Tumor Microenvironment ,medicine ,Humans ,030304 developmental biology ,Oncolytic Virotherapy ,0303 health sciences ,Tumor microenvironment ,business.industry ,Cancer ,Immunotherapy ,021001 nanoscience & nanotechnology ,medicine.disease ,Oncolytic virus ,Oncolytic Viruses ,Pharmaceutical Preparations ,Drug delivery ,Cancer cell ,Cancer research ,0210 nano-technology ,business - Abstract
Cancer therapies aim to kill tumor cells directly or engage the immune system to fight malignancy. Checkpoint inhibitors, oncolytic viruses, cell-based immunotherapies, cytokines, and adjuvants have been applied to prompt the immune system to recognize and attack cancer cells. However, systemic exposure of cancer therapies can induce unwanted adverse events. Intratumoral administration of potent therapies utilizes small amounts of drugs, in an effort to minimize systemic exposure and off-target toxicities. Here, we discuss the properties of the tumor microenvironment and transport considerations for intratumoral drug delivery. Specifically, we consider various tumor tissue factors and physicochemical factors that can affect tumor retention after intratumoral injection. We also review approved and clinical-stage intratumoral therapies and consider how the molecular and biophysical properties (e.g. size and charge) of these therapies influences intratumoral transport (e.g. tumor retention and cellular uptake). Finally, we offer a critical review and highlight several emerging approaches to promote tumor retention and limit systemic exposure of potent intratumoral therapies.
- Published
- 2020
- Full Text
- View/download PDF
22. Targeting IL-13Rα2 for effective treatment of malignant peripheral nerve sheath tumors in mouse models
- Author
-
James R. Connor, A. B. Madhankumar, Alexandre J. Bourcier, Christine Mau, Darren Wolfe, Elias Rizk, Oliver D. Mrowczynski, Kimberly Harbaugh, Becky Slagle-Webb, Stephan B. Abramson, Russell Payne, and Ganesh Shenoy
- Subjects
0303 health sciences ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Intratumoral Therapy ,General Medicine ,medicine.disease ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Toxicity ,Cancer research ,Medicine ,Pseudomonas exotoxin ,Cytotoxic T cell ,business ,Receptor ,030304 developmental biology - Abstract
OBJECTIVEMalignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft tissue sarcomas that harbor a high potential for metastasis and have a devastating prognosis. Combination chemoradiation aids in tumor control and decreases tumor recurrence but causes deleterious side effects and does not extend long-term survival. An effective treatment with limited toxicity and enhanced efficacy is critical for patients suffering from MPNSTs.METHODSThe authors recently identified that interleukin-13 receptor alpha 2 (IL-13Rα2) is overexpressed on MPNSTs and could serve as a precision-based target for delivery of chemotherapeutic agents. In the work reported here, a recombinant fusion molecule consisting of a mutant human IL-13 targeting moiety and a point mutant variant of Pseudomonas exotoxin A (IL-13.E13 K-PE4E) was utilized to treat MPNST in vitro in cell culture and in an in vivo murine model.RESULTSIL-13.E13 K-PE4E had a potent cytotoxic effect on MPNST cells in vitro. Furthermore, intratumoral administration of IL-13.E13 K-PE4E to orthotopically implanted MPNSTs decreased tumor burden 6-fold and 11-fold in late-stage and early-stage MPNST models, respectively. IL-13.E13 K-PE4E treatment also increased survival by 23 days in the early-stage MPNST model.CONCLUSIONSThe current MPNST treatment paradigm consists of 3 prongs: surgery, chemotherapy, and radiation, none of which, either singly or in combination, are curative or extend survival to a clinically meaningful degree. The results presented here provide the possibility of intratumoral therapy with a potent and highly tumor-specific cytotoxin as a fourth treatment prong with the potential to yield improved outcomes in patients with MPNSTs.
- Published
- 2019
- Full Text
- View/download PDF
23. In situ tumor vaccination with nanoparticle co-delivering CpG and STAT3 siRNA to effectively induce whole-body antitumor immune response
- Author
-
Ngoc Ha Hoang, Amanda W. Lund, Wassana Yantasee, Ruijie Wang, Shiuh Wen Luoh, Gordon B. Mills, Sancy A. Leachman, Worapol Ngamcherdtrakul, Joe W. Gray, Molly A. Nelson, Husam Y. Zaidan, Daniel S. Bejan, Moataz Reda, and Ryan S. Lane
- Subjects
Materials science ,medicine.medical_treatment ,Intratumoral Therapy ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Article ,Mice ,Immune system ,Cancer immunotherapy ,medicine ,Animals ,General Materials Science ,RNA, Small Interfering ,Mechanical Engineering ,Melanoma ,Vaccination ,Investigational New Drug ,Immunotherapy ,021001 nanoscience & nanotechnology ,medicine.disease ,0104 chemical sciences ,CpG site ,Mechanics of Materials ,Cancer research ,Nanoparticles ,0210 nano-technology - Abstract
The success of immunotherapy with immune checkpoint inhibitors (ICIs) in a subset of individuals has been very exciting. However, in many cancers, responses to current ICIs are modest and are seen only in a small subsets of patients. Herein, a widely applicable approach that increases the benefit of ICIs is reported. Intratumoral administration of augmenting immune response and inhibiting suppressive environment of tumors-AIRISE-02 nanotherapeutic that co-delivers CpG and STAT3 siRNA-results in not only regression of the injected tumor, but also tumors at distant sites in multiple tumor model systems. In particular, three doses of AIRISE-02 in combination with systemic ICIs completely cure both treated and untreated aggressive melanoma tumors in 63% of mice, while ICIs alone do not cure any mice. A long-term memory immune effect is also reported. AIRISE-02 is effective in breast and colon tumor models as well. Lastly, AIRISE-02 is well tolerated in mice and nonhuman primates. This approach combines multiple therapeutic agents into a single nanoconstruct to create whole-body immune responses across multiple cancer types. Being a local therapeutic, AIRISE-02 circumvents regulatory challenges of systemic nanoparticle delivery, facilitating rapid translation to the clinic. AIRISE-02 is under investigational new drug (IND)-enabling studies, and clinical trials will soon follow.
- Published
- 2021
24. Current status of intratumoral therapy for glioblastoma.
- Author
-
Mehta, Ankit, Linninger, Andreas, Lesniak, Maciej, and Engelhard, Herbert
- Abstract
With emerging drug delivery technologies becoming accessible, more options are expected to become available to patients with glioblastoma (GBM) in the near future. It is important for clinicians to be familiar with the underlying mechanisms and limitations of intratumoral drug delivery, and direction of recent research efforts. Tumor-adjacent brain is an extremely complex living matrix that creates challenges with normal tissue intertwining with tumor cells. For convection-enhanced delivery (CED), the role of tissue anisotropy for better predicting the biodistribution of the infusate has recently been studied. Computational predictive methods are now available to better plan CED therapy. Catheter design and placement-in addition to the agent being used-are critical components of any protocol. This paper overviews intratumoral therapies for GBM, highlighting key anatomic and physiologic perspectives, selected agents (especially immunotoxins), and some new developments such as the description of the glymphatic system. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
25. Advances in Targeting Cutaneous Melanoma
- Author
-
Laura Susok, Thilo Gambichler, Dimitri Kasakovski, and Marina Skrygan
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Population ,Review ,Targeted therapy ,combination therapy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,melanoma ,education ,RC254-282 ,intratumoral therapy ,education.field_of_study ,skin cancer ,business.industry ,Melanoma ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,targeted therapy ,Immune checkpoint ,Oncolytic virus ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Cancer research ,cancer therapy ,immunotherapy ,Skin cancer ,business - Abstract
Simple Summary Cutaneous Melanoma (CM), arising from pigment-producing melanocytes in the skin, is an aggressive cancer with high metastatic potential. While cutaneous melanoma represents only a fraction of all skin cancers (
- Published
- 2021
26. Image-guided intratumoral immunotherapy: Developing a clinically practical technology.
- Author
-
Som, Avik, Rosenboom, Jan-Georg, Chandler, Alana, Sheth, Rahul A., and Wehrenberg-Klee, Eric
- Subjects
- *
POLYMERIC drug delivery systems , *CONTROLLED release drugs , *IMMUNOTHERAPY - Abstract
[Display omitted] • Intratumoral immunotherapy is a rapidly growing clinical field bridging clinical image guided delivery by interventional radiology and controlled release drug delivery. • Real-time imaging feedback is necessary to ensure proper intratumoral delivery. • A variety of imaging modalities are under scientific exploration, yet only a few have the potential for near-future application in clinical settings, particularly CT and ultrasound. • Eventual clearance of the imaging contrast agent is ideal to minimize impact on interpretation of subsequent staging studies. • Controlled-release formulations will improve clinical adoption by minimizing the need for multiple procedures and may enhance efficacy of intratumoral immunoadjuvants. Immunotherapy has revolutionized the contemporary oncology landscape, with durable responses possible across a range of cancer types. However, the majority of cancer patients do not respond to immunotherapy due to numerous immunosuppressive barriers. Efforts to overcome these barriers and increase systemic immunotherapy efficacy have sparked interest in the local intratumoral delivery of immune stimulants to activate the local immune response and subsequently drive systemic tumor immunity. While clinical evaluation of many therapeutic candidates is ongoing, development is hindered by a lack of imaging confirmation of local delivery, insufficient intratumoral drug distribution, and a need for repeated injections. The use of polymeric drug delivery systems, which have been widely used as platforms for both image guidance and controlled drug release, holds promise for delivery of intratumoral immunoadjuvants and the development of an in situ cancer vaccine for patients with metastatic cancer. In this review, we explore the current state of the field for intratumoral delivery and methods for optimizing controlled drug release, as well as practical considerations for drug delivery design to be optimized for clinical image guided delivery particularly by CT and ultrasound. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
27. Intratumoral plasmid IL-12 expands CD8(+) T cells and induces a CXCR3 gene signature in triple-negative breast tumors that sensitizes patients to anti-PD-1 therapy
- Author
-
Erica Browning, Erika J. Crosby, Takuya Osada, Bernard A. Fox, Carlo Bifulco, Jendy Sell, Irene Wapnir, Melinda L. Telli, Kaitlin Zablotsky, Reneta Hermiz, Kellie Malloy Foerter, Robert H. Pierce, Chaitanya R. Acharya, Kim Jaffe, Donna Bannavong, Mai Hope Le, H. Kim Lyerly, Shawn M. Jensen, Carmen Ballesteros-Merino, David A. Canton, Christopher G. Twitty, Hiroshi Nagata, and Lauren Svenson
- Subjects
0301 basic medicine ,Cancer Research ,Receptors, CXCR3 ,Intratumoral Therapy ,Antigen presentation ,Triple Negative Breast Neoplasms ,CD8-Positive T-Lymphocytes ,Injections, Intralesional ,Article ,Immunophenotyping ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Breast cancer ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,Medicine ,Cytotoxic T cell ,Animals ,Humans ,Immune Checkpoint Inhibitors ,Melanoma ,business.industry ,Disease Management ,Gene signature ,medicine.disease ,Interleukin-12 ,Disease Models, Animal ,030104 developmental biology ,Electroporation ,Treatment Outcome ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Interleukin 12 ,Female ,business ,CD8 ,Iron Compounds ,Plasmids - Abstract
Purpose: Triple-negative breast cancer (TNBC) is an aggressive disease with limited therapeutic options. Antibodies targeting programmed cell death protein 1 (PD-1)/PD-1 ligand 1 (PD-L1) have entered the therapeutic landscape in TNBC, but only a minority of patients benefit. A way to reliably enhance immunogenicity, T-cell infiltration, and predict responsiveness is critically needed. Patients and Methods: Using mouse models of TNBC, we evaluate immune activation and tumor targeting of intratumoral IL12 plasmid followed by electroporation (tavokinogene telseplasmid; Tavo). We further present a single-arm, prospective clinical trial of Tavo monotherapy in patients with treatment refractory, advanced TNBC (OMS-I140). Finally, we expand these findings using publicly available breast cancer and melanoma datasets. Results: Single-cell RNA sequencing of murine tumors identified a CXCR3 gene signature (CXCR3-GS) following Tavo treatment associated with enhanced antigen presentation, T-cell infiltration and expansion, and PD-1/PD-L1 expression. Assessment of pretreatment and posttreatment tissue from patients confirms enrichment of this CXCR3-GS in tumors from patients that exhibited an enhancement of CD8+ T-cell infiltration following treatment. One patient, previously unresponsive to anti–PD-L1 therapy, but who exhibited an increased CXCR3-GS after Tavo treatment, went on to receive additional anti–PD-1 therapy as their immediate next treatment after OMS-I140, and demonstrated a significant clinical response. Conclusions: These data show a safe, effective intratumoral therapy that can enhance antigen presentation and recruit CD8 T cells, which are required for the antitumor efficacy. We identify a Tavo treatment-related gene signature associated with improved outcomes and conversion of nonresponsive tumors, potentially even beyond TNBC.
- Published
- 2021
28. Case Report: Microfragmented Adipose Tissue Drug Delivery in Canine Mesothelioma: A Case Report on Safety, Feasibility, and Clinical Findings
- Author
-
Offer Zeira, Erica Ghezzi, Letizia Pettinari, Valentina Re, Davide M. Lupi, Silvia L. Benali, Simone Borgonovo, Giulio Alessandri, Francesco Petrella, Rita Paroni, Michele Dei Cas, Carlo Tremolada, Valentina Coccè, and Augusto Pessina
- Subjects
Drug ,040301 veterinary sciences ,Intratumoral Therapy ,media_common.quotation_subject ,Adipose tissue ,Case Report ,Pharmacology ,0403 veterinary science ,03 medical and health sciences ,paclitaxel ,Pharmacokinetics ,Medicine ,Mesothelioma ,030304 developmental biology ,media_common ,0303 health sciences ,lcsh:Veterinary medicine ,General Veterinary ,business.industry ,04 agricultural and veterinary sciences ,medicine.disease ,adipose tissue ,mesothelioma ,Drug delivery ,Toxicity ,dog ,drug delivery ,Peritoneal mesothelioma ,lcsh:SF600-1100 ,Veterinary Science ,business - Abstract
Mesothelioma is a rare lethal tumor of dogs and humans involving cavities of the body. Dogs are considered a model for new drugs and therapeutic methods since they present spontaneous diseases similar to humans. Microfragmented adipose tissue (MFAT) uploaded by paclitaxel (PTX) is a drug delivery medium providing slow release of chemotherapic drugs. A dog affected by pleural, pericardial, and peritoneal mesothelioma was treated by 17 intracavitary ultrasound-guided injections of MFAT-PTX over 22 months. A long-lasting improvement of general conditions was observed, treatment was well-tolerated, and no toxicity or hypersensitivity was reported. Pharmacokinetic (PK) data indicated low drug localization in the circulatory system and a tendency to enter or remain in the extravascular compartments of the body. Indeed, low levels of free-circulating drugs for a short time produced low toxicity, whereas, a higher intracavitary PTX concentration can have major pharmacological efficacy. To our knowledge, this is the first time that mesothelioma has been treated using such a procedure, and this should be considered as a novel therapeutic approach. The low systemic absorption suggests the possible role of MFAT-PTX for loco-regional/intratumoral therapy also useful in other types of tumors, and further investigation is warranted.
- Published
- 2021
29. An experimental model of anti-PD-1 resistance exhibits activation of TGFß and Notch pathways and is sensitive to local mRNA immunotherapy
- Author
-
Hui Cao, Kerstin A. Heyl, Tatiana Tolstykh, Hui Qu, Marie Bernardo, Timothy R. Wagenaar, Kuldeep Singh, Katie Malley, Emma Wang, Errin Roberts, Yu-an Zhang, Fangxian Sun, Jack Pollard, Sue Ryan, Dinesh S. Bangari, Eladio J. Márquez, Dmitri Wiederschain, Joon Sang Lee, Keith Bahjat, and Natalia Malkova
- Subjects
0301 basic medicine ,Cell type ,checkpoint blockade resistance ,medicine.medical_treatment ,Immunology ,Notch signaling pathway ,Biology ,syngeneic tumor model ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,Neoplasms ,medicine ,Animals ,Humans ,Immunology and Allergy ,RNA, Messenger ,intratumoral therapy ,RC254-282 ,Original Research ,Antigen Presentation ,cancer immunotherapy ,Antigen processing ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,RC581-607 ,Immune checkpoint ,Blockade ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunologic diseases. Allergy ,Signal transduction ,Research Article - Abstract
Immune checkpoint blockade elicits durable anti-cancer responses in the clinic, however a large proportion of patients do not benefit from treatment. Several mechanisms of innate and acquired resistance to checkpoint blockade have been defined and include mutations of MHC I and IFNγ signaling pathways. However, such mutations occur in a low frequency of patients and additional mechanisms have yet to be elucidated. In an effort to better understand acquired resistance to checkpoint blockade, we generated a mouse tumor model exhibiting in vivo resistance to anti-PD-1 antibody treatment. MC38 tumors acquired resistance to PD-1 blockade following serial in vivo passaging. Lack of sensitivity to PD-1 blockade was not attributed to dysregulation of PD-L1 or β2M expression, as both were expressed at similar levels in parental and resistant cells. Similarly, IFNγ signaling and antigen processing and presentation pathways were functional in both parental and resistant cell lines. Unbiased gene expression analysis was used to further characterize potential resistance mechanisms. RNA-sequencing revealed substantial differences in global gene expression, with tumors resistant to anti-PD-1 displaying a marked reduction in expression of immune-related genes relative to parental MC38 tumors. Indeed, resistant tumors exhibited reduced immune infiltration across multiple cell types, including T and NK cells. Pathway analysis revealed activation of TGFβ and Notch signaling in anti-PD-1 resistant tumors, and activation of these pathways was associated with poorer survival in human cancer patients. While pharmacological inhibition of TGFβ and Notch in combination with PD-1 blockade decelerated tumor growth, a local mRNA-based immunotherapy potently induced regression of resistant tumors, resulting in complete tumor remission, and resensitized tumors to treatment with anti-PD-1. Overall, this study describes a novel anti-PD-1 resistant mouse tumor model and underscores the role of two well-defined signaling pathways in response to immune checkpoint blockade. Furthermore, our data highlights the potential of intratumoral mRNA therapy in overcoming acquired resistance to PD-1 blockade.
- Published
- 2021
- Full Text
- View/download PDF
30. 799 Durable responses and immune activation with intratumoral electroporation of pIL-12 plus pembrolizumab in actively progressing anti-PD-1 refractory advanced melanoma: KEYNOTE 695 interim data
- Author
-
Christopher Stuart Baker, Jack Lee, Tom Van Hagen, Victoria Atkinson, Bernard A. Fox, Carmen Ballesteros-Merino, Eric D. Whitman, Kellie Malloy Foerter, Reneta Hermiz, Sajeve Thomas, Scott J. Diede, Elizabeth Buchbinder, Katy K. Tsai, Catalin Mihalcioiu, Shawn M. Jensen, Rachel Roberts-Thomson, Sandra Aung, David A. Canton, Alain Algazi, Christopher G. Twitty, Mecker G. Möller, Clemens Krepler, Donna Bannavong, Emmett V. Schmidt, Marcus O. Butler, John R. Hyngstrom, Erica Browning, Jon Salazar, M. Shaheen, Adil Daud, Matteo S. Carlino, Andrew Mant, C. Lance Cowey, Gregory A. Daniels, Lauren Svenson, Pablo Fernandez-Penas, Igor Puzanov, Jendy Sell, and Andrew Haydon
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Melanoma ,Intratumoral Therapy ,Ipilimumab ,Pembrolizumab ,medicine.disease ,Interim analysis ,Lesion ,Internal medicine ,medicine ,Nivolumab ,medicine.symptom ,Adverse effect ,business ,medicine.drug - Abstract
Background Electroporated plasmid IL-12 (TAVO or tavokinogene telseplasmid) is a novel pro-inflammatory intratumoral therapy with substantial single agent activity in melanoma, which has been shown to synergize with anti-PD-1 antibodies in patients predicted as non-responders to anti-PD-1.1 2 Interim data from patients with stage III/IV melanoma actively progressing on anti-PD-1 antibody are presented herein. Methods Patients with confirmed disease progression by RECIST v1.1 after at least 12 weeks of treatment on pembrolizumab or nivolumab (or combination checkpoint blockade) and within 12 weeks of last dose (with no intervening therapies) were enrolled. There was no limit on the number of prior lines of therapy. At least one accessible lesion was electroporated with plasmid IL-12 (pIL-12-EP) on days 1, 5 and 8 every 6 weeks and pembrolizumab was administered every 3 weeks. Tumor response in treated and untreated lesions was assessed by RECIST v1.1 every 12 weeks. Endpoints include ORR, safety, PFS, OS, and DOR. Results The first 56 patients treated of 100 planned were included in this interim analysis. Of these, 84% had Stage IV disease, 30% had M1c or M1d disease, and 27% had prior exposure to ipilimumab. In 54 efficacy evaluable patients the investigator-assessed ORR was 30% (3 CR/13 PR), 5 patients had 100% reduction of target lesions. All responses have been confirmed, only two responding patient progressed while on study, 2 patients completed the study with ongoing responses (figures 1 and 2). In patients with M1c/M1d disease, the ORR was 35.2% (n=6/17). Tumor reduction was observed in untreated lesions in 12 of 12 patients who had unaccessible lesions or accessible untreated lesions. The median overall survival (mOS) and duration of response (mDOR) has not been reached, with a median follow-up time of 13 months. Grade 3 treatment-related adverse events (TRAEs) were seen in 5.4% of patients, and there were no grade 4/5 TRAEs. The rate of grade 3 treatment-emergent (TEAEs) regardless of cause was 23.2%. The median time for pIL-12-EP treatment was 10 minutes (range 2,46). Consistent with prior studies of single-agent pIL-12-EP, tumor IHC, and transcriptomic assessments revealed hallmarks of antigen-specific antitumor immunity in this study. Additional analyses including microbiome, TCR clonality, and peripheral blood biomarker assays will be presented. Conclusions In this rigorously defined PD-1 antibody refractory patient population, the addition of pIL-12-EP to PD-1 antibody therapy induced deep, durable, systemic response in local treated and distant visceral metastatic untreated lesions with nominal systemic toxicity. Trial Registration Trial Registration: NCT#03132675 Ethics Approval The study was approved by a central IRB and/or local institutional IRBs/Ethics Committees as required for each participating institution. Consent Written informed consent was obtained from the patients participating within the trial, the current abstract does not contain sensitive or identifiable information requiring an additional consent from patients. References Algazi A, Bhatia S, Agarwala S, et al. Intratumoral delivery of tavokinogene telseplasmid yields systemic immune responses in metastatic melanoma patients. Annals of Oncology 2019;31:532–540. Algazi A, Twitty C, Tsai K, et al. Phase II trial for IL-12 plasmid transfection and PD-1 blockade in immunologically quiescent melanoma. Clinical Cancer Research 2020;26:2827-2837.
- Published
- 2020
- Full Text
- View/download PDF
31. Toll-Like Receptor 4 Expression on Lymphoma Cells Is Critical for Therapeutic Activity of Intratumoral Therapy With Synthetic TLR4 Agonist Glucopyranosyl Lipid A
- Author
-
Jan ter Meulen, Hailing Lu, Alec Betancur, and Michael Chen
- Subjects
0301 basic medicine ,Agonist ,Cancer Research ,medicine.drug_class ,Intratumoral Therapy ,Antigen presentation ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,immune system diseases ,hemic and lymphatic diseases ,medicine ,tumor microenvironment ,TLR4 ,B-cell lymphoma ,Original Research ,Tumor microenvironment ,Chemistry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Lymphoma ,030104 developmental biology ,Oncology ,intratumoral treatment ,030220 oncology & carcinogenesis ,Cancer research ,GLA ,CD8 - Abstract
Intratumoral (IT) injections of Glucopyranosyl lipid A (G100), a synthetic toll-like receptor 4 (TLR4) agonist formulated in a stable emulsion, resulted in T-cell inflammation of the tumor microenvironment (TME) and complete cure of 60% of mice with large established A20 lymphomas. Strong abscopal effects on un-injected lesions were observed in a bilateral tumor model and surviving mice resisted a secondary tumor challenge. Depletion of CD8 T-cells, but not CD4 or NK cells, abrogated the anti-tumor effect. Unexpectedly, TLR4 knock-out rendered A20 tumors completely non-responsive to G100. In vitro studies showed that GLA has direct effect on A20 cells, but not on A20 cells deficient for TLR4. As shown by genotyping and phenotyping analysis, G100 strongly activated antigen presentation functions in A20 cells in vitro and in vivo and induced their apoptosis in a dose dependent manner. Similarly, the TLR4 positive human mantle cell lymphoma line Mino showed in vitro activation with G100 that was blocked with an anti-TLR4 antibody. In the A20 model, direct activation of B-lymphoma cells with G100 is sufficient to induce protective CD8 T-cell responses and TLR4 expressing human B-cell lymphomas may be amenable to this therapy as well.
- Published
- 2020
32. Constructing a Biomaterial to Simulate Extracellular Drug Transport in Solid Tumors
- Author
-
Aric Huang, Cory Berkland, M. Laird Forrest, Chad Groer, Jimmy Y. Song, Samuel R. Crowl, and Sebastian G. Huayamares
- Subjects
Polymers and Plastics ,Extracellular transport ,Intratumoral Therapy ,Drug Compounding ,Bioengineering ,Biocompatible Materials ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Polyethylene Glycols ,Biomaterials ,chemistry.chemical_compound ,Mice ,Drug Delivery Systems ,Cell Line, Tumor ,Hyaluronic acid ,PEG ratio ,Materials Chemistry ,Extracellular ,Animals ,Humans ,Hyaluronic Acid ,Drug Carriers ,technology, industry, and agriculture ,Hydrogels ,Glatiramer Acetate ,021001 nanoscience & nanotechnology ,Xenograft Model Antitumor Assays ,0104 chemical sciences ,chemistry ,Head and Neck Neoplasms ,Drug delivery ,Self-healing hydrogels ,Cancer research ,0210 nano-technology ,Ex vivo ,Biotechnology - Abstract
Designing an in vitro model of the tumor extracellular microenvironment to screen intratumoral drugs is an active challenge. As recent clinical successes of human intratumoral therapies stimulate research on intratumoral delivery, a need for a 3D tumor model to screen intratumoral therapies arises. When injecting the drug formulation directly into the tumor, the biophysics affecting intratumoral retention must be considered; especially for biologic therapies, which may be dominated by extracellular transport mechanisms. Fibrotic regions in solid tumors are typically rich in collagen I fibers. Using shear rheology, head and neck tumors with higher collagen density show a higher stiffness. Similarly, the stiffness of the hyaluronic acid (HA) hydrogel models is increased by adding collagen fibers to model the bulk biomechanical properties of solid tumors. HA hydrogels are then used as intratumoral injection site simulators to model in vitro the retention of glatiramer acetate (GA) and polyethylene glycol (PEG) administered intratumorally. Both compounds are also injected in murine tumors and retention is studied ex vivo for comparison. Retention of GA in the hydrogels is significantly longer than PEG, analogous to the solid tumors, suggesting the utility of HA hydrogels with collagen I fibers for screening extracellular drug transport after intratumoral administration.
- Published
- 2020
33. Lysis-independent potentiation of immune checkpoint blockade by oncolytic virus
- Author
-
Jedd D. Wolchok, Jacob Ricca, Brian Ko, Taha Merghoub, Mathieu Gigoux, Anton Oseledchyk, Tyler Walther, Gopa Iyer, Cailian Liu, Dmitriy Zamarin, and Gil Redelman-Sidi
- Subjects
0301 basic medicine ,animal structures ,medicine.medical_treatment ,T cell ,Intratumoral Therapy ,animal diseases ,viruses ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,NDV ,PD-1 ,Medicine ,lysis ,business.industry ,Cancer ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Immune checkpoint ,3. Good health ,Oncolytic virus ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunogenic cell death ,bladder cancer ,immunotherapy ,business ,Research Paper - Abstract
Intratumoral therapy with oncolytic viruses is increasingly being explored as a strategy to potentiate an immune response against cancer, but it remains unknown whether such therapy should be restricted to cancers sensitive to virus-mediated lysis. Using Newcastle Disease Virus (NDV) as a model, we explore immunogenic potential of an oncolytic virus in bladder cancer, where existing immunotherapy with PD-1 and PD-L1-targeting antibodies to date has shown suboptimal response rates. Infection of human and mouse bladder cancer cells with NDV resulted in immunogenic cell death, activation of innate immune pathways, and upregulation of MHC and PD-L1 in all tested cell lines, including the cell lines completely resistant to NDV-mediated lysis. In a bilateral flank NDV-lysis-resistant syngeneic murine bladder cancer model, intratumoral therapy with NDV led to an increase of immune infiltration in both treated and distant tumors and a shift from an inhibitory to effector T cell phenotype. Consequently, combination of intratumoral NDV with systemic PD-1 or CTLA-4 blockade led to improved local and abscopal tumor control and overall survival. These findings encourage future clinical trials combining intratumoral NDV therapy with systemic immunomodulatory agents and underscore the rationale for such treatments irrespective of tumor cell sensitivity to NDV-mediated lysis.
- Published
- 2018
34. Tumor immunotherapy using adenovirus vaccines in combination with intratumoral doses of CpG ODN.
- Author
-
Geary, S., Lemke, C., Lubaroff, D., and Salem, A.
- Subjects
- *
CANCER immunotherapy , *TUMOR immunology , *ADENOVIRUSES , *VIRAL vaccines , *LABORATORY mice , *CELL receptors , *IMMUNOLOGICAL adjuvants , *T cells - Abstract
The combination of viral vaccination with intratumoral (IT) administration of CpG ODNs is yet to be investigated as an immunotherapeutic treatment for solid tumors. Here, we show that such a treatment regime can benefit survival of tumor-challenged mice. C57BL/6 mice bearing ovalbumin (OVA)-expressing EG.7 thymoma tumors were therapeutically vaccinated with adenovirus type 5 encoding OVA (Ad5-OVA), and the tumors subsequently injected with the immunostimulatory TLR9 agonist, CpG-B ODN 1826 (CpG), 4, 7, 10, and 13 days later. This therapeutic combination resulted in enhanced mean survival times that were more than 3.5× longer than naïve mice, and greater than 40% of mice were cured and capable of resisting subsequent tumor challenge. This suggests that an adaptive immune response was generated. Both Ad5-OVA and Ad5-OVA + CpG IT treatments led to significantly increased levels of H-2 K-OVA-specific CD8+ lymphocytes in the peripheral blood and intratumorally. Lymphocyte depletion studies performed in vivo implicated both NK cells and CD8+ lymphocytes as co-contributors to the therapeutic effect. Analysis of tumor infiltrating lymphocytes (TILs) on day 12 post-tumor challenge revealed that mice treated with Ad5-OVA + CpG IT possessed a significantly reduced percentage of regulatory T lymphocytes (Tregs) within the CD4+ lymphocyte population, compared with TILs isolated from mice treated with Ad5-OVA only. In addition, the proportion of CD8+ TILs that were OVA-specific was reproducibly higher in the mice treated with Ad5-OVA + CpG IT compared with other treatment groups. These findings highlight the therapeutic potential of combining intratumoral CpG and vaccination with virus encoding tumor antigen. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
35. An open-label, single-arm pilot study of EUS-guided brachytherapy with phosphorus-32 microparticles in combination with gemcitabine +/- nab-paclitaxel in unresectable locally advanced pancreatic cancer (OncoPaC-1): Technical details and study protocol
- Author
-
Franklin C. Wong, Nicholas N. Nissen, Matthew H.G. Katz, Manoop S. Bhutani, R. Tuli, Simon K. Lo, Jason B. Klapman, David R. Fogelman, Ashish Soman, Gauri R. Varadhachary, Andrew Eugene Hendifar, Beth Chasen, Ben S. Singh, Sarah E. Hoffe, Robert A. Wolff, William D. Erwin, Ghassan El-Haddad, Rutika Mehta, Irina M. Cazacu, Eric P. Tamm, Joseph M. Herman, and Eugene J. Koay
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Intratumoral Therapy ,Brachytherapy ,pancreatic cancer ,chemotherapy ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Clinical Research ,Pancreatic cancer ,medicine ,Radiology, Nuclear Medicine and imaging ,endoscopy ,EUS ,intratumoral therapy ,radiotherapy ,Cancer ,Chemotherapy ,Hepatology ,medicine.diagnostic_test ,business.industry ,Gastroenterology ,Evaluation of treatments and therapeutic interventions ,fine needle injection ,medicine.disease ,Gemcitabine ,Locally advanced pancreatic cancer ,Endoscopy ,Radiation therapy ,neoplasia ,Orphan Drug ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,030211 gastroenterology & hepatology ,Radiology ,Digestive Diseases ,business ,Rapid Communication ,phosphorus-32 ,medicine.drug - Abstract
Current treatment options for patients with unresectable locally advanced pancreatic cancer (LAPC) include chemotherapy alone or followed by chemoradiation or stereotactic body radiotherapy. However, the prognosis for these patients remains poor, with a median overall survival
- Published
- 2019
36. Intratumoral and Combination Therapy in Melanoma and Other Skin Cancers
- Author
-
Adil Daud, Alain Algazi, and Arielle Oglesby
- Subjects
Oncology ,medicine.medical_specialty ,Skin Neoplasms ,Combination therapy ,Intratumoral Therapy ,Dermatology ,Injections, Intralesional ,Radiosurgery ,Cancer Vaccines ,T-Lymphocytes, Regulatory ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Pharmacotherapy ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,Internal medicine ,medicine ,Tumor Microenvironment ,Humans ,Progression-free survival ,Melanoma ,Tumor microenvironment ,business.industry ,Cancer ,General Medicine ,medicine.disease ,Neoadjuvant Therapy ,Progression-Free Survival ,Disease Progression ,Administration, Intravenous ,Immunotherapy ,Skin cancer ,Neoplasm Recurrence, Local ,business ,T-Lymphocytes, Cytotoxic - Abstract
Skin cancer, as the most physically accessible malignancy, allows for the greatest variety in treatment innovation. The last 2 decades have seen striking increases in the life expectancies of those diagnosed with malignant melanoma. However, many cases remain in which disease prevails against standard treatment, and those patients rely on continuing ingenuity. Drugs that can be injected directly into patients’ tumors have become increasingly promising, not least for the reduction in side effects observed. Intratumoral therapy encompasses a wide array of agents, from chemotherapeutic drugs to cancer vaccines. While each show some efficacy, those agents which regulate the immune system likely have the greatest potential for preventing disease progression or recurrence. Recent research has highlighted the importance of the presence of cytotoxic T cells and of keeping regulatory T cells in check. Thus, manipulating the tumor microenvironment is a need in skin cancer therapy, which intratumoral delivery can potentially address. In order to find the best approach to each person’s disease, more studies are needed to test intralesional agents in combination with currently approved therapies and with each other.
- Published
- 2019
37. In situ antitumor vaccination: Targeting the tumor microenvironment
- Author
-
Hanwen Li, Bin Shao, Xiawei Wei, Jiayun Yu, and Yongyao Wu
- Subjects
0301 basic medicine ,Physiology ,Intratumoral Therapy ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,Clinical Biochemistry ,Cancer Vaccines ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Tumor Microenvironment ,Humans ,Tumor microenvironment ,biology ,business.industry ,Immunogenicity ,Cell Biology ,Immunotherapy ,Combined Modality Therapy ,Vaccination ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Immunization ,Antibody ,business - Abstract
Tumor microenvironment is known to play important roles in tumor progression. Many therapies, targeting the tumor microenvironment, are designed and applied in the clinic. One of these approaches is in situ antitumor therapy. This way, bacteria, antibodies, plasmid DNA, viruses, and cells are intratumorally delivered into the tumor site as "in-situ antitumor vaccine," which seeks to enhance immunogenicity and generate systemic T cell responses. In addition, this intratumoral therapy can alter the tumor microenvironment from immunosuppressive to immunostimulatory while limiting the risk of systemic exposure and associated toxicity. Contemporarily, promising preclinical results and some initial success in clinical trials have been obtained after intratumoral therapy.
- Published
- 2019
38. Tumor Therapy: In Situ Tumor Vaccination with Nanoparticle Co‐Delivering CpG and STAT3 siRNA to Effectively Induce Whole‐Body Antitumor Immune Response (Adv. Mater. 31/2021)
- Author
-
Wassana Yantasee, Molly A. Nelson, Joe W. Gray, Husam Y. Zaidan, Sancy A. Leachman, Worapol Ngamcherdtrakul, Gordon B. Mills, Moataz Reda, Daniel S. Bejan, Ryan S. Lane, Ngoc Ha Hoang, Amanda W. Lund, Shiuh-Wen Luoh, and Ruijie Wang
- Subjects
Materials science ,biology ,Mechanical Engineering ,medicine.medical_treatment ,Intratumoral Therapy ,Melanoma ,Tumor therapy ,medicine.disease ,Vaccination ,Immune system ,Cancer immunotherapy ,CpG site ,Mechanics of Materials ,Cancer research ,medicine ,biology.protein ,General Materials Science ,STAT3 - Published
- 2021
- Full Text
- View/download PDF
39. Abstract CT240: LUMINOS-101: Phase 2 study of PVSRIPO with pembrolizumab in recurrent glioblastoma
- Author
-
Michael C. Brown, Andrea Kelly, Lori Mixson, Lisa Franklin, Shirley Ong, Nicholas Butowski, William T. Curry, Prakash Ambady, Annick Desjardins, Patrick Y. Wen, Andrew E Sloan, Henry S. Friedman, Robert Cavaliere, Dina Randazzo, Matthias Gromeier, LeAnn Jackson, and Garrett Nichols
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,Intratumoral Therapy ,medicine.medical_treatment ,Phases of clinical research ,Immunotherapy ,Pembrolizumab ,medicine.disease ,Radiation therapy ,Tolerability ,Internal medicine ,Glioma ,medicine ,business - Abstract
Background: The prognosis for patients (pts) with recurrent (r) glioblastoma (GBM) is poor, with no highly effective approved therapies. Treatment failure may result from poor penetration of drugs through the blood-brain barrier and the immunosuppressive nature of the tumor microenvironment (TME). PVSRIPO, a recombinant poliovirus (PV):rhinovirus chimera, is a novel, non-neurovirulent, intratumoral immunotherapy. Trial results in rGBM pts showed greater long-term survival with PVSRIPO monotherapy (21%, 36-60 months [mos]) vs. criteria-matched external controls (4%, 36 mos; 2%, 60 mos; Desjardins 2018 NEJM). PVSRIPO targets CD155 (PV receptor), expressed on solid tumors and APC. PVSRIPO IT infection results in inflammatory-mediated destruction of tumor cells but non-lethal lingering infection in TME APC. This leads to a type-I/III interferon-dominant inflammation and ultimately, tumor antigen-specific T cell activation and recruitment (Brown 2017 Sci Transl Med), which is potentiated by immunologic recall to intratumoral replicating virus via prior vaccination. Induction of type-1 IFN dominant inflammation and compensatory activation of the PD-1:PD-L1 immune checkpoint (IC) pathway support investigation of PVSRIPO in combination with PD-1/L1 IC inhibitors. Immunologically cold mouse glioma models showed PVSRIPO+anti-PD-1 therapy resulted in greater anti-tumor response than either agent. Trial design/objectives: LUMINOS-101 (NCT04479241) is a phase 2, multicenter, open-label, single-arm study of intratumoral infusion of PVSRIPO (day 1: 5×107 TCID50) followed by the anti-PD-1 monoclonal antibody pembrolizumab (200mg IV q3w) in adult pts with rGBM. The trial objective is to evaluate the anti-tumor activity, safety, and tolerability of the combination. Eligibility criteria: Pts ≥18 years who had prior PV and boost IPOL® immunizations, histologically confirmed supratentorial rGBM, infusible 1 to ≤5.5cm enhancing disease, confirmed disease progression following prior therapies, and KPS ≥70 will be enrolled. Key exclusion criteria include multifocal disease; discontinuation of prior anti-PD-1/L1 agent for toxicity; prior intratumoral therapy, immunotherapy, or radiotherapy within 12 weeks; high-dose systemic corticosteroids; chemotherapy, anti-VEGF, or TTF therapy ≤1-6 weeks depending on the therapy; serious cerebral herniation syndrome; extensive leptomeningeal, subependymal, or ≥1cm enhancing disease crossing the midline; and severe active comorbidities. Endpoints: Primary endpoints are objective response rate, duration of response, and safety. Secondary endpoints include overall and progression-free survival and disease control rate and duration. Exploratory endpoints include assessment of tumor and blood for biomarkers of response. The study is currently enrolling in the US, and results will inform the design of a randomized phase 3 trial. Citation Format: Andrea True Kelly, Prakash Ambady, Michael Brown, Nicholas Butowski, Robert Cavaliere, William Curry, Annick Desjardins, Lisa Franklin, Henry Friedman, Matthias Gromeier, LeAnn Jackson, Lori Mixson, Shirley Ong, Dina Randazzo, Andrew Sloan, Patrick Wen, Garrett Nichols. LUMINOS-101: Phase 2 study of PVSRIPO with pembrolizumab in recurrent glioblastoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT240.
- Published
- 2021
- Full Text
- View/download PDF
40. ItRECIST adapted efficacy assessment in solid tumors treated with intratumoral immunotherapy
- Author
-
A. Hernando-Calvo, Maria Vieito, Elena Elez, Xavier Serres-Creixams, Gemma Mur-Bonet, Vladimir Galvao, Teresa Macarulla, Josep Tabernero, Mafalda Oliveira, Omar Saavedra Santa Gadea, Guillem Cunill-Macià, Guzman Alonso, Honey Kumar Oberoi, Enriqueta Felip, Iosune Baraibar, Eva Muñoz-Couselo, Ignacio Matos, Marc Diez, Elena Garralda, and Irene Brana
- Subjects
Cancer Research ,Oncology ,business.industry ,Intratumoral Therapy ,Immune checkpoint inhibitors ,medicine.medical_treatment ,medicine ,Cancer research ,Immunotherapy ,business - Abstract
2557 Background: The development of human intratumoral therapy (HIT-IT) has surged as a promising strategy to overcome resistance to checkpoint inhibitors (CPI), promoting a stronger tumor-specific immune response while reducing systemic exposure. A broad variety of agents (i.e: oncolytic viruses, toll-like receptors agonists) administered both in superficial- and deep-seated lesions are being currently tested in clinical trials (CT). Due to the local intervention on tumors, radiological assessment by standard RECIST is challenging and new methods of response that capture and integrate the local and systemic response to HIT-IT are needed. We aimed to evaluate the feasibility and clinical utility of itRECIST (Goldmacher et al., 2020) in patients (pts) treated with HIT-IT in early phase CT. Methods: Retrospective analysis of a cohort of pts with different solid tumor types enrolled in CT including HIT-IT in our institution between August’18 and January’21. Clinical characteristics were collected. Efficacy in target-injected (T-I) and target-non-injected (T-NI) lesions was assessed by objective response rate (ORR) and disease control rate (DCR), as per itRECIST. Overall disease ORR and DCR were assessed per RECIST 1.1/iRECIST. Treatment-related adverse events (TRAEs) were assessed with CTCAE v.5.0. ORR was calculated with Clopper-Pearson method. Survival analysis was made using Kaplan-Meier method. Results: A total of 37 pts were included. Median age was 66 years, 19 pts (51%) were male, all pts had ECOG 0-1. 24 pts (65%) were CPI-naïve. Median previous lines of therapy was 2 (range [r]: 0-11). All pts (100%) received minimum 1 dose of HIT-IT. 6 pts (16%) were treated with monotherapy and 31 pts (84%) in combination with CPI. Median HIT-IT and CPI doses administered were 4 (r: 1-9) and 2 (r: 1-13), respectively. Injected lesions: cutaneous (16.2%), subcutaneous (21.6%), lymph node (32.4%), liver (29.7%). Median size of T-I lesions was 40 mm (r: 19-260). At data cutoff, 32 pts were evaluable. Median follow-up was 14.4 weeks (r: 1.0-81.1). Per RECIST 1.1, overall ORR was 6% (95% CI, 5-7) and DCR was 38% (95% CI, 21-56). Per itRECIST, ORR was 19% (95% CI, 7-36) and DCR was 63% (95% CI, 44-79) in T-I lesions (n = 32), and 10% (95% CI, 22-27) and 48% (95% CI, 29-67) in T-NI lesions (n = 29). Mean decrease in responding T-I and T-NI lesions was -47% (r: -21 to -100) and -41% (r: -26 to -59), respectively. No non-target (NT) lesion was injected. Median progression-free survival was 7.4 weeks (95% CI, 6.6 – 8.2). Median overall survival was 10.0 months (95% CI, 2.3 – 17.7). Incidence of TRAE was 58% (grade 1-2 IT-related pyrexia 43%; grade 3-4, 5%). No treatment-related deaths were recorded. Conclusions: ItRECIST is feasible to implement and adds precision to the radiological assessment of local and distant anti-tumor activity of HIT-IT. No safety issues were detected in our cohort.
- Published
- 2021
- Full Text
- View/download PDF
41. Intratumoral modulation of the inducible co-stimulator ICOS by recombinant oncolytic virus promotes systemic anti-tumour immunity
- Author
-
Rikke B. Holmgaard, Peter Palese, Dmitriy Zamarin, Padmanee Sharma, Taha Merghoub, Jacob Ricca, James P. Allison, Tamar Plitt, and Jedd D. Wolchok
- Subjects
0301 basic medicine ,Intratumoral Therapy ,animal diseases ,viruses ,Science ,Newcastle disease virus ,General Physics and Astronomy ,chemical and pharmacologic phenomena ,Biology ,Lymphocyte Activation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Immunomodulation ,Inducible T-Cell Co-Stimulator Protein ,Inducible T-Cell Co-Stimulator Ligand ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,Cell Line, Tumor ,Animals ,Humans ,CTLA-4 Antigen ,Mice, Knockout ,Oncolytic Virotherapy ,Recombination, Genetic ,Multidisciplinary ,Innate immune system ,Neoplasms, Experimental ,General Chemistry ,biochemical phenomena, metabolism, and nutrition ,Immune checkpoint ,3. Good health ,Oncolytic virus ,Blockade ,Mice, Inbred C57BL ,ICOS LIGAND ,Oncolytic Viruses ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology - Abstract
Emerging data suggest that locoregional cancer therapeutic approaches with oncolytic viruses can lead to systemic anti-tumour immunity, although the appropriate targets for intratumoral immunomodulation using this strategy are not known. Here we find that intratumoral therapy with Newcastle disease virus (NDV), in addition to the activation of innate immunity, upregulates the expression of T-cell co-stimulatory receptors, with the inducible co-stimulator (ICOS) being most notable. To explore ICOS as a direct target in the tumour, we engineered a recombinant NDV-expressing ICOS ligand (NDV-ICOSL). In the bilateral flank tumour models, intratumoral administration of NDV-ICOSL results in enhanced infiltration with activated T cells in both virus-injected and distant tumours, and leads to effective rejection of both tumours when used in combination with systemic CTLA-4 blockade. These findings highlight that intratumoral immunomodulation with an oncolytic virus expressing a rationally selected ligand can be an effective strategy to drive systemic efficacy of immune checkpoint blockade., Oncolytic viruses induce a variety of immune targets in the infected tumours. Here, the authors show that Newcastle Disease Virus (NDV) upregulates the inducible co-stimulator (ICOS) on T cells and that intratumoral targeting of ICOS with engineered NDV in combination with CTLA-4 blockade induces systemic anti-tumour immunity in mice.
- Published
- 2017
42. Endoscopic ultrasound guided interventions in the management of pancreatic cancer.
- Author
-
Kerdsirichairat T and Shin EJ
- Abstract
There has been a growing interest in developing endoscopic ultrasound (EUS)-guided interventions for pancreatic cancer, some of which have become standard of care. There are two main factors that drive these advancements to facilitate treatment of patients with pancreatic cancer, ranging from direct locoregional therapy to palliation of symptoms related to inoperable pancreatic cancer. Firstly, an upper EUS has the capability to access the entire pancreas-lesions in the pancreatic head and uncinate process can be accessed from the duodenum, and lesions in the pancreatic body and tail can be accessed from the stomach. Secondly, there has been a robust development of devices that allow through-the-needle interventions, such as placement of fiducial markers, brachytherapy, intratumoral injection, gastroenterostomy creation, and ablation. While these techniques are rapidly emerging, data from a multicenter randomized controlled trial for some procedures are awaited prior to their adoption in clinical settings., Competing Interests: Conflict-of-interest statement: No conflict of interest related to the manuscript., (©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
43. Commentary: Current status of intratumoral therapy for glioblastoma
- Author
-
Ankit Mehta
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Intratumoral Therapy ,Medicine ,Current (fluid) ,business ,medicine.disease ,Glioblastoma - Published
- 2016
- Full Text
- View/download PDF
44. Talimogene Laherparepvec combined with anti-PD-1 based immunotherapy for unresectable stage III-IV melanoma: a case series
- Author
-
Pauline Funchain, Patricia Rayman, Lillian D. Sun, Michael J. McNamara, Charles S. Tannenbaum, Brian R. Gastman, Jung Min Song, C. Marcela Diaz-Montero, and Jennifer S. Ko
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Intratumoral Therapy ,Immunology ,Ipilimumab ,Herpesvirus 1, Human ,Pembrolizumab ,lcsh:RC254-282 ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Stage IIIC ,Melanoma ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Pharmacology ,Biological Products ,business.industry ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Oncolytic virus ,030104 developmental biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Immunotherapy ,Nivolumab ,business ,Talimogene laherparepvec ,Research Article ,medicine.drug - Abstract
Background Talimogene Laherparepvec (T-VEC) is an oncolytic virus approved as an intratumoral therapy for treating unresectable stage IIIB-IV metastatic melanoma. The mechanisms of action for T-VEC and checkpoint inhibitor are highly complementary. Recent studies have shown that combining checkpoint inhibitor therapy with T-VEC injection can lead to improved response rates for stage IIIB-IV melanoma patients. Methods We reviewed 10 consecutive cases of stage IIIC to stage IVM1b melanoma patients that received T-VEC plus checkpoint inhibitor(s) therapy (pembrolizumab, ipilimumab/nivolumab, or nivolumab) treated between June 2016 and August 2017 at the Cleveland Clinic with a median follow-up of 7 months (range: 4 to 13 months). Responses of injected (on-target) and uninjected (off-target) lesions were evaluated according to RECIST 2.0. Results The overall response rate for on-target lesions was 90%, with 6 patients experiencing a complete response in injected lesions. Two patients had off-target lesions, which were completely resolved after treatment. Blood samples were tested for 3 complete responders and 2 partial responders. CD4:CD8 ratio and frequencies of circulating PD1+ CD4 and CD8 T cells were elevated in complete responders but not partial responders. One patient died due to causes unrelated to melanoma and one patient died of progression of the disease. Conclusion Our data suggest that combining checkpoint inhibitor(s) with T-VEC injection may provide a synergistic efficacy for patients with unresectable melanoma. We observed a better overall response rate and complete response rate compared to published studies on similar therapeutic regimens.
- Published
- 2018
- Full Text
- View/download PDF
45. Intralesional Therapy for In-transit and Satellite Metastases in Melanoma
- Author
-
Michael J. Mastrangelo and Kendra J. Feeney
- Subjects
Skin Neoplasms ,biology ,business.industry ,Intratumoral Therapy ,Melanoma ,Cancer ,Antineoplastic Agents ,Injections, Intralesional ,Bacille Calmette Guerin ,biology.organism_classification ,medicine.disease ,Adjuvants, Immunologic ,Oncology ,Immunology ,Cancer research ,Humans ,Medicine ,Surgery ,Satellite (biology) ,Neoplasm Metastasis ,business - Abstract
Intratumoral therapy with bacteria/bacterial products dates to at least the 1890s. Over the decades this has expanded beyond the use of microbes and microbial products to include chemicals, cancer chemotherapeutic agents, cytokines, recombinant organisms, and hybrid molecules. The appeal of this method of delivery is the ability to deliver high concentrations of the therapeutic agent directly to the tumor, often with minimal side effects. This article summarizes the use and efficacy of the various agents used in the past and present in the treatment of in-transit and satellite metastases in melanoma.
- Published
- 2015
- Full Text
- View/download PDF
46. Advances in Targeting Cutaneous Melanoma.
- Author
-
Kasakovski, Dimitri, Skrygan, Marina, Gambichler, Thilo, Susok, Laura, and Ricci, Francesca
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *DISEASE progression , *VIRUSES , *IMMUNE checkpoint inhibitors , *MELANOMA , *CANCER chemotherapy , *PATIENT-centered care , *SKIN tumors , *IMMUNOTHERAPY , *THERAPEUTICS - Abstract
Simple Summary: Cutaneous Melanoma (CM), arising from pigment-producing melanocytes in the skin, is an aggressive cancer with high metastatic potential. While cutaneous melanoma represents only a fraction of all skin cancers (<5%), it accounts for most skin-cancer-related deaths worldwide. Immune checkpoint inhibition has been the first therapeutic approach to significantly benefit patient survival after treatment. Nevertheless, the immunosuppressive tumor microenvironment and the intrinsic and acquired treatment resistance of melanoma remain crucial challenges. Combining local and systemic treatment offers the potential to augment therapeutic response and overcome resistance, although, complex drug combinations can harbor an increased risk of immune-related adverse events. The aim of this review is to give current insight into studies combining systemic and local therapeutic approaches to overcome drug resistance, prime melanoma cells for therapy, and improve overall treatment response in CM patients. To date, the skin remains the most common cancer site among Caucasians in the western world. The complex, layered structure of human skin harbors a heterogenous population of specialized cells. Each cell type residing in the skin potentially gives rise to a variety of cancers, including non-melanoma skin cancer, sarcoma, and cutaneous melanoma. Cutaneous melanoma is known to exacerbate and metastasize if not detected at an early stage, with mutant melanomas tending to acquire treatment resistance over time. The intricacy of melanoma thus necessitates diverse and patient-centered targeted treatment options. In addition to classical treatment through surgical intervention and radio- or chemotherapy, several systemic and intratumoral immunomodulators, pharmacological agents (e.g., targeted therapies), and oncolytic viruses are trialed or have been recently approved. Moreover, utilizing combinations of immune checkpoint blockade with targeted, oncolytic, or anti-angiogenic approaches for patients with advanced disease progression are promising approaches currently under pre-clinical and clinical investigation. In this review, we summarize the current 'state-of-the-art' as well as discuss emerging agents and regimens in cutaneous melanoma treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
47. Intratumoral Therapy II: In Vitro and In Vivo Immunologic Testing and Therapy Options
- Author
-
Max H. Cohen, Ronald B. Herberman, and Alfred S. Ketcham
- Subjects
Oncology ,medicine.medical_specialty ,Melanoma-associated antigen ,business.industry ,Melanoma ,Intratumoral Therapy ,medicine.disease ,law.invention ,Breast cancer ,Immune system ,medicine.anatomical_structure ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Lung cancer ,business ,Lymph node - Abstract
In a prospective randomized trial in patients with metastatic melanoma we compared two agents that had been used for metastatic melanoma intratumoral injections. Each patient had progressive metastatic disease no longer surgically controllable. Multiple metastases included satellitosis in the form of progressive nodules around the previously excised original melanoma site, and/or in-transit metastases in the form of observable tumor nodules progressing in a linear fashion toward a lymph node bearing area. As patients were receiving intratumoral injections, serially collected blood samples were tested for general immunologic reactivity and anti-melanoma reactivity. Specificity controls included breast cancer and lung cancer extracts. Additionally, as a measure of cell-mediated immunity, the patients were serially skin tested against antigens to measure general and melanoma-specific immunity. Depending on the patients’ clinical courses, we have divided the patients retrospectively into groups whose clinical courses were either better or worse than their cohorts, and determined the relationship between the immune testing and the clinical courses that the patients were experiencing as the serial testing was being conducted. Additionally, in a group of similar patients that were ‘cured for life’, we analyze their treatment in light of therapeutic attempts made by others to similarly haptenize melanoma antigens. We describe potential synergies between newly discovered melanoma therapies and intratumoral injection treatments and point out potential combination therapies that may offer the potential for enhancement of antitumor effects without increased systemic toxicity, a desirable goal now that combinations of recently acceptable immunotherapies have been associated with severe potential toxicities including death.
- Published
- 2018
- Full Text
- View/download PDF
48. Intratumoral Therapy I: Association of Immunotherapy with Permanent Long Term Cure of Metastatic Cancer
- Author
-
Alfred S. Ketcham, Max H. Cohen, and Ronald B. Herberman
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Intratumoral Therapy ,Melanoma ,Cancer ,Immunotherapy ,Disease ,medicine.disease ,medicine.anatomical_structure ,Internal medicine ,Scalp ,medicine ,Forehead ,Prospective cohort study ,business - Abstract
We have demonstrated in a randomized prospective study the superiority of intratumoral (intralesional) dinitrochlorobenzene (DNCB) compared to intratumoral (intralesional) bacillus Calmette-Guerin (BCG) in the treatment of progressive metastatic melanoma. The metastatic melanoma was in the form of satellitosis and/or in-transit metastases. We now demonstrate the ability of intralesional DNCB to permanently cure a selected group of patients with the same clinical criteria, and describe their clinical characteristics and treatment regimens. The described cured patients were followed for their remaining lifetimes, for up to 30 years, after being immunotherapeutically rendered free of metastatic cancer. They were each female, between the ages of 51 and 56 when intratumoral treatments were begun. Treatment was for progressive cutaneous and subcutaneous metastatic disease in the leg in two cases and for rapidly spreading scalp and forehead metastases in another. In each case the disease was not surgically controllable. Treatments were continued for 6 to 26 months. Subsequently the patients survived tumor free for either 18 years, dying at age 83; or for 24 years, dying at age 89; or for 30 years, dying at age 97. There has been a significant interest in recent years in immunotherapy for advanced cancer. The absence of systemic toxicity in the cured patients reported herein provides a basis for consideration of combining intratumoral treatments with current effective but systemically more toxic immunotherapeutic approaches to some metastatic cancers. Intratumoral treatments may be particularly applicable in cases of uncontrollable melanoma with cutaneous metastases, without evidence of distant spread, as in the current report.
- Published
- 2018
- Full Text
- View/download PDF
49. Endoscopic ultrasound.guided fine-needle tissue acquisition – A review and update of literature
- Author
-
Bronte A. Holt, Benjamin Tharian, Robert H. Hawes, Shyam Varadarajulu, Shantel Hebert-Magee, Muhammad K. Hasan, Nayana Elizabeth George, Udayakumar Navaneethan, Konrad Krall, and Ashley Canipe
- Subjects
Endoscopic ultrasound ,medicine.medical_specialty ,Gastrointestinal tumors ,molecular markers ,Intratumoral Therapy ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,medicine ,In patient ,fine-needle aspiration ,lcsh:RC799-869 ,Neoadjuvant therapy ,General Environmental Science ,training ,medicine.diagnostic_test ,business.industry ,rapid onsite cytologic evaluation ,tissue acquisition ,Tissue acquisition ,Fine-needle aspiration ,030220 oncology & carcinogenesis ,endoscopic ultrasound ,cytology ,Tissue diagnosis ,030211 gastroenterology & hepatology ,lcsh:Diseases of the digestive system. Gastroenterology ,Radiology ,business ,fine needle core biopsy - Abstract
Ever since the first endoscopic ultrasound-guided fine needle aspiration (FNA) was done in 1992, the procedure has evolved to become an indispensable tool for tissue acquisition in patients with gastrointestinal tumors and periluminal lesions. With the growing evidence of neoadjuvant therapy and research into intratumoral therapy, the need to obtain tissue diagnosis for tumors is quite apparent. This review provides an overall perspective to the endosonographer on various issues that are a key for best practices in FNA, in addition to being an update for practicing experienced endosonographers.
- Published
- 2015
- Full Text
- View/download PDF
50. Development of an adaptive electroporation system for intratumoral plasmid DNA delivery
- Author
-
Arya J. Bahrami, Anandaroop Mukhopadhyay, Richard J. Connolly, Douglas W. Brown, Robert H. Pierce, David A. Canton, and Jean S. Campbell
- Subjects
0301 basic medicine ,Genetic enhancement ,Intratumoral Therapy ,Cell ,Biophysics ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Electrochemistry ,medicine ,Animals ,Viability assay ,Physical and Theoretical Chemistry ,Reporter gene ,Mice, Inbred BALB C ,Electroporation ,Gene Transfer Techniques ,General Medicine ,Transfection ,DNA ,Equipment Design ,Genetic Therapy ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Genetically Engineered Mouse ,Cancer research ,Female ,Plasmids - Abstract
Intratumoral electroporation of plasmid DNA encoding the proinflammatory cytokine interleukin 12 promotes innate and adaptive immune responses correlating with anti-tumor effects. Clinical electroporation conditions are fixed parameters optimized in preclinical tumors, which consist of cells implanted into skin. These conditions have little translatability to clinically relevant tumors, as implanted models cannot capture the heterogeneity encountered in genetically engineered mouse models or clinical tumors. Variables affecting treatment outcome include tumor size, degree of vascularization, fibrosis, and necrosis, which can result in suboptimal gene transfer and variable therapeutic outcomes. To address this, a feedback controlled electroporation generator was developed, which is capable of assessing the electrochemical properties of tissue in real time. Determination of these properties is accomplished by impedance spectroscopy and equivalent circuit model parameter estimation. Model parameters that estimate electrical properties of cell membranes are used to adjust electroporation parameters for each applied pulse. Studies performed in syngeneic colon carcinoma tumors (MC38) and spontaneous mammary tumors (MMTV-PyVT) demonstrated feedback-based electroporation is capable of achieving maximum expression of reporter genes with significantly less variability and applied energy. These findings represent an advancement to the practice of gene electro-transfer, as reducing variability and retaining transfected cell viability is paramount to treatment success.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.