1
|
Rastogi S, Joshi A, Sato N, Lee S, Lee MJ, Trepel JB, Neckers L. An update on the status of HSP90 inhibitors in cancer clinical trials. Cell Stress Chaperones 2024; 29:519-539. [PMID: 38878853 PMCID: PMC11260857 DOI: 10.1016/j.cstres.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
The evolutionary conserved molecular chaperone heat shock protein 90 (HSP90) plays an indispensable role in tumorigenesis by stabilizing client oncoproteins. Although the functionality of HSP90 is tightly regulated, cancer cells exhibit a unique dependence on this chaperone, leading to its overexpression, which has been associated with poor prognosis in certain malignancies. While various strategies targeting heat shock proteins (HSPs) involved in carcinogenesis have been explored, only inhibition of HSP90 has consistently and effectively resulted in proteasomal degradation of its client proteins. To date, a total of 22 HSP90 inhibitors (HSP90i) have been tested in 186 cancer clinical trials, as reported by clinicaltrials.gov. Among these trials, 60 % have been completed, 10 % are currently active, and 30 % have been suspended, terminated, or withdrawn. HSP90 inhibitors (HSP90i) have been used as single agents or in combination with other drugs for the treatment of various cancer types in clinical trials. Notably, improved clinical outcomes have been observed when HSP90i are used in combination therapies, as they exhibit a synergistic antitumor effect. However, as single agents, HSP90i have shown limited clinical activity due to drug-related toxicity or therapy resistance. Recently, active trials conducted in Japan evaluating TAS-116 (pimitespib) have demonstrated promising results with low toxicity as monotherapy and in combination with the immune checkpoint inhibitor nivolumab. Exploratory biomarker analyses performed in various trials have demonstrated target engagement that suggests the potential for identifying patient populations that may respond favorably to the therapy. In this review, we discuss the advances made in the past 5 years regarding HSP90i and their implications in anticancer therapeutics. Our focus lies in evaluating drug efficacy, prognosis forecast, pharmacodynamic biomarkers, and clinical outcomes reported in published trials. Through this comprehensive review, we aim to shed light on the progress and potential of HSP90i as promising therapeutic agents in cancer treatment.
Collapse
Affiliation(s)
- Shraddha Rastogi
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD, USA
| | - Abhinav Joshi
- Urologic Oncology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD, USA
| | - Nahoko Sato
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD, USA
| | - Sunmin Lee
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD, USA
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD, USA
| | - Jane B Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD, USA
| | - Len Neckers
- Urologic Oncology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Sweeney R, Omstead AN, Fitzpatrick JT, Zheng P, Gorbunova A, Grayhack EE, Goel A, Khan AF, Kosovec JE, Wagner PL, Jobe BA, Kelly RJ, Zaidi AH. Sitravatinib combined with PD-1 blockade enhances cytotoxic T-cell infiltration by M2 to M1 tumor macrophage repolarization in esophageal adenocarcinoma. Carcinogenesis 2024; 45:210-219. [PMID: 38019590 DOI: 10.1093/carcin/bgad087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is a leading cause of cancer-related mortality. Sitravatinib is a novel multi-gene tyrosine kinase inhibitor (TKI) that targets tumor-associated macrophage (TAM) receptors, VEGF, PDGF and c-Kit. Currently, sitravatinib is actively being studied in clinical trials across solid tumors and other TKIs have shown efficacy in combination with immune checkpoint inhibitors (ICI) in cancer models. In this study, we investigated the anti-tumor activity of sitravatinib alone and in combination with PD-1 blockade in an EAC rat model. Treatment response was evaluated by mortality, pre- and post-treatment MRI, gene expression, immunofluorescence and immunohistochemistry. Our results demonstrated adequate safety and significant tumor shrinkage in animals treated with sitravatinib, and more profoundly, sitravatinib and PD-1 inhibitor, AUNP-12 (P < 0.01). Suppression of TAM receptors resulted in increased gene expression of pro-inflammatory cytokines and decreased expression of anti-inflammatory cytokines, enhanced infiltration of CD8+ T cells, and M2 to M1 macrophage phenotype repolarization in the tumor microenvironment of treated animals (P < 0.01). Moreover, endpoint immunohistochemistry staining corroborated the anti-tumor activity by downregulation of Ki67 and upregulation of Caspase-3 in the treated animals. Additionally, pretreatment gene expression of TAM receptors and PD-L1 were significantly higher in major responders compared with the non-responders, in animals that received sitravatinib and AUNP-12 (P < 0.02), confirming that TAM suppression enhances the efficacy of PD-1 blockade. In conclusion, this study proposes a promising immunomodulatory strategy using a multi-gene TKI to overcome developed resistance to an ICI in EAC, establishing rationale for future clinical development.
Collapse
Affiliation(s)
- Ryan Sweeney
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Ashten N Omstead
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - John T Fitzpatrick
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Ping Zheng
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Anastasia Gorbunova
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Erin E Grayhack
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Arul Goel
- University of California Santa Barbara, Santa Barbara, CA, USA
| | - Alisha F Khan
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | | | - Patrick L Wagner
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Blair A Jobe
- Allegheny Health Network, Esophageal Institute, Pittsburgh, PA, USA
| | - Ronan J Kelly
- Baylor University Medical Center at Dallas, Charles A. Sammons Cancer Center, Dallas, TX, USA
| | - Ali H Zaidi
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Omstead AN, Paskewicz M, Gorbunova A, Zheng P, Salvitti MS, Mansoor R, Reed P, Ballengee S, Wagner PL, Jobe BA, Kelly RJ, Zaidi AH. CSF-1R inhibitor, pexidartinib, sensitizes esophageal adenocarcinoma to PD-1 immune checkpoint blockade in a rat model. Carcinogenesis 2022; 43:842-850. [PMID: 35552655 DOI: 10.1093/carcin/bgac043] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/14/2022] [Accepted: 05/10/2022] [Indexed: 11/14/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is a leading cause of cancer deaths. Pexidartinib, a multi-gene tyrosine kinase inhibitor, through targeting CSF-1R, down modulates macrophage mediated pro-survival tumor signaling. Previously, CSF-1R inhibitors have successfully shown to enhance antitumor activity of PD-1/PD-L1 inhibitors by suppressing tumor immune evasion, in solid tumors. In this study, we investigated the antitumor activity of pexidartinib alone or in combination with blockade of PD-1 in a de novo EAC rat model. Here, we showed limited toxicity with significant tumor shrinkage in pexidartinib treated animals compared to controls, single agent and in combination with a PD-1 inhibitor, AUNP-12. Suppression of CSF-1/CSF-1R axis resulted in enhanced infiltration of CD3+CD8+ T cells with reduced M2 macrophage polarization, in the tumor microenvironment (TME). Endpoint tissue gene expression in pexidartinib treated animals demonstrated upregulation of BAX, Cas3, TNFα, IFNγ and IL6 and downregulation of Ki67, IL13, IL10, TGFβ and Arg1 (p<0.05). Additionally, among the pexidartinib treated animals responders compared to non-responders demonstrated a significant upregulation of pre-treatment CSF-1 gene, confirming that tumor associated macrophage suppression directly translates to clinical benefit. Moreover, a post-treatment serum cytokine assay exhibited similar systemic trends as the gene expression in the TME, depicting increases in pro-inflammatory cytokines and decreases in anti-inflammatory cytokines. In conclusion, our study established a promising combinatorial strategy using a CSF-1R inhibitor to overcome resistance to PD-1/PD-L1 axis blockade in an EAC model, providing the rationale for future clinical strategies.
Collapse
Affiliation(s)
- Ashten N Omstead
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Michael Paskewicz
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Anastasia Gorbunova
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Ping Zheng
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Madison S Salvitti
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Rubab Mansoor
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Payton Reed
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Sydne Ballengee
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Patrick L Wagner
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Blair A Jobe
- Allegheny Health Network, Esophageal Institute, Pittsburgh, PA, USA
| | - Ronan J Kelly
- Baylor University Medical Center at Dallas, Department of Hematology and Oncology, Dallas, TX, USA
| | - Ali H Zaidi
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Abstract
Background Ocular adverse events are common dose-limiting toxicities in cancer patients treated with HSP90 inhibitors, such as AUY922; however, the pathology and molecular mechanisms that mediate AUY922-induced retinal toxicity remain undescribed. Methods The impact of AUY922 on mouse retinas and cell lines was comprehensively investigated using isobaric tags for relative and absolute quantitation (iTRAQ)‑based proteomic profiling and pathway enrichment analysis, immunohistochemistry and immunofluorescence staining, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, MTT assay, colony formation assay, and western blot analysis. The effect of AUY922 on the Transient Receptor Potential cation channel subfamily M member 1 (TRPM1)-HSP90 chaperone complex was characterized by coimmunoprecipitation. TRPM1-regulated gene expression was analyzed by RNAseq analysis and gene set enrichment analysis (GSEA). The role of TRPM1 was assessed using both loss-of-function and gain-of-function approaches. Results Here, we show that the treatment with AUY922 induced retinal damage and cell apoptosis, dysregulated the photoreceptor and retinal pigment epithelium (RPE) layers, and reduced TRPM1 expression. Proteomic profiling and functional annotation of differentially expressed proteins reveals that those related to stress responses, protein folding processes, regulation of apoptosis, cell cycle and growth, reactive oxygen species (ROS) response, cell junction assembly and adhesion regulation, and proton transmembrane transport were significantly enriched in AUY922-treated cells. We found that AUY922 triggered caspase-3-dependent cell apoptosis, increased ROS production and inhibited cell growth. We determined that TRPM1 is a bona fide HSP90 client and characterized that AUY922 may reduce TRPM1 expression by disrupting the CDC37-HSP90 chaperone complex. Additionally, GSEA revealed that TRPM1-regulated genes were associated with retinal morphogenesis in camera-type eyes and the JAK-STAT cascade. Finally, gain-of-function and loss-of-function analyses validated the finding that TRPM1 mediated the cell apoptosis, ROS production and growth inhibition induced by AUY922. Conclusions Our study demonstrates the pathology of AUY922-induced retinal toxicity in vivo. TRPM1 is an HSP90 client, regulates photoreceptor morphology and function, and mediates AUY922-induced cytotoxicity. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-021-00751-5.
Collapse
|
5
|
Epp-Ducharme B, Dunne M, Fan L, Evans JC, Ahmed L, Bannigan P, Allen C. Heat-activated nanomedicine formulation improves the anticancer potential of the HSP90 inhibitor luminespib in vitro. Sci Rep 2021; 11:11103. [PMID: 34045581 PMCID: PMC8160139 DOI: 10.1038/s41598-021-90585-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 01/06/2023] Open
Abstract
The heat shock protein 90 inhibitor, luminespib, has demonstrated potent preclinical activity against numerous cancers. However, clinical translation has been impeded by dose-limiting toxicities that have necessitated dosing schedules which have reduced therapeutic efficacy. As such, luminespib is a prime candidate for reformulation using advanced drug delivery strategies that improve tumor delivery efficiency and limit off-target side effects. Specifically, thermosensitive liposomes are proposed as a drug delivery strategy capable of delivering high concentrations of drug to the tumor in combination with other chemotherapeutic molecules. Indeed, this work establishes that luminespib exhibits synergistic activity in lung cancer in combination with standard of care drugs such as cisplatin and vinorelbine. While our research team has previously developed thermosensitive liposomes containing cisplatin or vinorelbine, this work presents the first liposomal formulation of luminespib. The physico-chemical properties and heat-triggered release of the formulation were characterized. Cytotoxicity assays were used to determine the optimal drug ratios for treatment of luminespib in combination with cisplatin or vinorelbine in non-small cell lung cancer cells. The formulation and drug combination work presented in this paper offer the potential for resuscitation of the clinical prospects of a promising anticancer agent.
Collapse
Affiliation(s)
| | - Michael Dunne
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Linyu Fan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - James C Evans
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Lubabah Ahmed
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
6
|
Zaidi AH, Kelly RJ, Gorbunova A, Omstead AN, Salvitti MS, Zheng P, Kosovec JE, Lee S, Ayazi S, Babar L, Finley GG, Goel A, Jobe BA. Intratumoral immunotherapy with STING agonist, ADU-S100, induces CD8+ T-cell mediated anti-tumor immunity in an esophageal adenocarcinoma model. Oncotarget 2021; 12:292-303. [PMID: 33659041 PMCID: PMC7899550 DOI: 10.18632/oncotarget.27886] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is a deadly disease with limited treatment options. STING is a transmembrane protein that activates transcription of interferon genes, resulting in stimulation of APCs and enhanced CD8+ T-cell infiltration. The present study evaluates STING agonists, alone and in combination with radiation to determine durable anticancer activity in solid tumors. MATERIALS AND METHODS Esophagojejunostomy was performed on rats to induce reflux leading to the development of EAC. At 32 weeks post operatively, rats received intratumorally either 50 μg STING (ADU-S100) or placebo (PBS), +/- 16Gy radiation. Drug activity was evaluated by pre- and post- treatment MRI, histology, immunofluorescence and RT-PCR. RESULTS Mean MRI tumor volume decreased by 30.1% and 50.8% in ADU-S100 and ADU-S100 + radiation animals and increased by 76.7% and 152.4% in placebo and placebo + radiation animals, respectively (P < 0.0001). Downstream gene expression, pre- to on- and post- treatment, demonstrated significant upregulation of IFNβ, TNFα, IL-6, and CCL-2 in the treatment groups vs. placebo. On- or post- treatment, radiation alone, ADU-S100 alone, and ADU-S100 + radiation groups demonstrated enhanced PD-LI expression, induced by upregulation of CD8+ T-cells (p < 0.01). CONCLUSIONS ADU-S100 +/- radiation exhibits potent antitumor activity and a promising immunomodulatory profile in a de novo EAC.
Collapse
Affiliation(s)
- Ali H. Zaidi
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
- Co-first authors and contributed equally to this work
| | - Ronan J. Kelly
- Department of Hematology and Oncology, Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Co-first authors and contributed equally to this work
| | - Anastasia Gorbunova
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ashten N. Omstead
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Madison S. Salvitti
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ping Zheng
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Juliann E. Kosovec
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Soyoung Lee
- Department of Radiation Oncology, Allegheny Health Network, Pittsburgh, PA, USA
| | - Shahin Ayazi
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Laila Babar
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Gene G. Finley
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Blair A. Jobe
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
7
|
Khalafi S, Lockhart AC, Livingstone AS, El-Rifai W. Targeted Molecular Therapies in the Treatment of Esophageal Adenocarcinoma, Are We There Yet? Cancers (Basel) 2020; 12:E3077. [PMID: 33105560 PMCID: PMC7690268 DOI: 10.3390/cancers12113077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023] Open
Abstract
Esophageal adenocarcinoma is one of the leading causes of cancer-related deaths worldwide. The incidence of esophageal adenocarcinoma has increased at an alarming rate in the Western world and long-term survival remains poor. Current treatment approaches involve a combination of surgery, chemotherapy, and radiotherapy. Unfortunately, standard first-line approaches are met with high rates of recurrence and metastasis. More recent investigations into the distinct molecular composition of these tumors have uncovered key genetic and epigenetic alterations involved in tumorigenesis and progression. These discoveries have driven the development of targeted therapeutic agents in esophageal adenocarcinoma. While many agents have been studied, therapeutics targeting the human epidermal growth factor receptor (HER2) and vascular endothelial growth factor (VEGF) pathways have demonstrated improved survival. More recent advances in immunotherapies have also demonstrated survival advantages with monoclonal antibodies targeting the programmed death ligand 1 (PD-L1). In this review we highlight recent advances of targeted therapies, specifically agents targeting receptor tyrosine kinases, small molecule kinase inhibitors, and immune checkpoint inhibitors. While targeted therapeutics and immunotherapies have significantly improved survival, the benefits are limited to patients whose tumors express biomarkers such as PD-L1 and HER2. Survival remains poor for the remainder of patients with esophageal adenocarcinoma, underscoring the critical need for development of novel treatment strategies.
Collapse
Affiliation(s)
- Shayan Khalafi
- Department of Surgery, Miler School of Medicine, University of Miami, Miami, FL 33136, USA; (S.K.); (A.S.L.)
| | - Albert Craig Lockhart
- Department of Medicine, Miler School of Medicine, University of Miami, Miami, FL 33136, USA;
- Sylvester Comprehensive Cancer Center, Miler School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Alan S. Livingstone
- Department of Surgery, Miler School of Medicine, University of Miami, Miami, FL 33136, USA; (S.K.); (A.S.L.)
| | - Wael El-Rifai
- Department of Surgery, Miler School of Medicine, University of Miami, Miami, FL 33136, USA; (S.K.); (A.S.L.)
- Department of Medicine, Miler School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL 33136, USA
| |
Collapse
|
8
|
Kelly RJ, Zaidi AH, Smith MA, Omstead AN, Kosovec JE, Matsui D, Martin SA, DiCarlo C, Werts ED, Silverman JF, Wang DH, Jobe BA. The Dynamic and Transient Immune Microenvironment in Locally Advanced Esophageal Adenocarcinoma Post Chemoradiation. Ann Surg 2019; 268:992-999. [PMID: 28806299 DOI: 10.1097/sla.0000000000002410] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The aim of this study was to assess the impact of chemoradiation on the immune microenvironment to influence and optimally design future neoadjuvant clinical trials. SUMMARY BACKGROUND DATA Programmed death (PD)-1 inhibitors in metastatic gastroesophageal cancer have demonstrated response rates of approximately 25% in programmed death ligand-1 (PD-L1+) tumors. Unfortunately, the majority of patients do not respond. Therefore, a rationale strategy of combining immunotherapeutic agents with chemoradiation in earlier stage esophageal cancer may prevent metastatic disease in patients. METHODS To determine the effects of chemoradiation on resected esophageal adenocarcinomas, we examined the immune microenvironment pre- and post-chemoradiation using immunohistochemistry, quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), and functional analysis of tumor-infiltrating lymphocytes. Additionally, to assess the duration and dependency of radiation-induced PD-L1 upregulation, a surgical rat reflux model of esophageal adenocarcinoma is used. First, tumor-bearing animals were dosed with single-fraction 13Gy or 16Gy radiation to determine safety, dose correlation, and PD-L1 upregulation using qRT-PCR post-radiation. Next, longitudinal PD-L1 expression levels within individual animals were determined using serial endoscopic biopsies at baseline, 1, 5, and 9 weeks post 16Gy radiation. RESULTS The majority of cancers displayed enhanced interferon γ and activated CD8+ T lymphocytes at the tumor stroma interface. These tumors also demonstrated enhanced upregulation of PD-L1 and multiple other immune checkpoints including TIM3, GITR, IDO1, LAG3, OX40, and KIR. The animal model results indicated PD-L1 upregulation is dose-dependent and transiently elevated post radiation exposure. CONCLUSIONS Collectively, these findings provide insights into the evolving immune landscape after chemoradiation and have significant implications for neoadjuvant trial designs that will combine radiotherapy with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ronan J Kelly
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD
| | - Ali H Zaidi
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Matthew A Smith
- Department of Pathology and Laboratory Medicine, Allegheny Health Network, Pittsburgh, PA
| | - Ashten N Omstead
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Juliann E Kosovec
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Daisuke Matsui
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Samantha A Martin
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Christina DiCarlo
- Department of Pathology and Laboratory Medicine, Allegheny Health Network, Pittsburgh, PA
| | - E Day Werts
- Division of Radiation Oncology, Allegheny Health Network, Pittsburgh, PA
| | - Jan F Silverman
- Department of Pathology and Laboratory Medicine, Allegheny Health Network, Pittsburgh, PA
| | - David H Wang
- Esophageal Diseases Center, VA North Texas Health Care System and University of Texas Southwestern Medical Center, Dallas, TX
| | - Blair A Jobe
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Augello G, Emma MR, Cusimano A, Azzolina A, Mongiovì S, Puleio R, Cassata G, Gulino A, Belmonte B, Gramignoli R, Strom SC, McCubrey JA, Montalto G, Cervello M. Targeting HSP90 with the small molecule inhibitor AUY922 (luminespib) as a treatment strategy against hepatocellular carcinoma. Int J Cancer 2018; 144:2613-2624. [DOI: 10.1002/ijc.31963] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Giuseppa Augello
- Institute of Biomedicine and Molecular Immunology “Alberto Monroy”National Research Council (CNR) Palermo Italy
| | - Maria Rita Emma
- Institute of Biomedicine and Molecular Immunology “Alberto Monroy”National Research Council (CNR) Palermo Italy
| | - Antonella Cusimano
- Institute of Biomedicine and Molecular Immunology “Alberto Monroy”National Research Council (CNR) Palermo Italy
| | - Antonina Azzolina
- Institute of Biomedicine and Molecular Immunology “Alberto Monroy”National Research Council (CNR) Palermo Italy
| | - Sarah Mongiovì
- Institute of Biomedicine and Molecular Immunology “Alberto Monroy”National Research Council (CNR) Palermo Italy
| | - Roberto Puleio
- Istituto Zooprofilattico Sperimentale della Sicilia “A. Mirri”Histopathology and Immunohistochemistry Laboratory Palermo Italy
| | - Giovanni Cassata
- Istituto Zooprofilattico Sperimentale della Sicilia “A. Mirri”Histopathology and Immunohistochemistry Laboratory Palermo Italy
| | - Alessandro Gulino
- Tumor Immunology Unit, Department of Health ScienceUniversity of Palermo Palermo Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health ScienceUniversity of Palermo Palermo Italy
| | - Roberto Gramignoli
- Division of Pathology, Department of Laboratory MedicineKarolinska Institutet Stockholm Sweden
| | - Stephen C. Strom
- Division of Pathology, Department of Laboratory MedicineKarolinska Institutet Stockholm Sweden
| | - James A. McCubrey
- Department of Microbiology and ImmunologyBrody School of Medicine at East Carolina University Greenville North Carolina USA
| | - Giuseppe Montalto
- Institute of Biomedicine and Molecular Immunology “Alberto Monroy”National Research Council (CNR) Palermo Italy
- Biomedic Department of Internal Medicine and SpecialtiesUniversity of Palermo Palermo Italy
| | - Melchiorre Cervello
- Institute of Biomedicine and Molecular Immunology “Alberto Monroy”National Research Council (CNR) Palermo Italy
| |
Collapse
|
10
|
Omstead AN, Kosovec JE, Matsui D, Martin SA, Smith MA, Aaron Guel D, Kolano J, Komatsu Y, Habib F, Lai C, Christopher K, Kelly RJ, Zaidi AH, Jobe BA. Serial Endoscopic Evaluation of Esophageal Disease in a Cancer Model: A Paradigm Shift for Esophageal Adenocarcinoma (EAC) Drug Discovery and Development. Cancer Invest 2018; 36:363-370. [PMID: 30142016 DOI: 10.1080/07357907.2018.1499029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A rat model of surgically induced reflux recapitulates the development and progression of human esophageal adenocarcinoma (EAC). In this study, reflux was induced in rats followed by postoperative endoscopy with biopsy, to diagnose and monitor disease progression. Overall, percentage agreement between visual endoscopy and gold standard histology was 95%, with disease-specific classification accuracies of 100% and 75% for Barrett's with dysplasia and EAC, respectively. Additionally, the percentage agreement for biopsy in tumors >4 mm was 75%. Thereby, establishing endoscopic evaluation as a reliable tool to assess disease progression and provide biopsies for downstream correlates in a de novo EAC model.
Collapse
Affiliation(s)
- Ashten N Omstead
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Juliann E Kosovec
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Daisuke Matsui
- b Department of Gastroenterological Surgery , Kanazawa University Hospital , Kanazawa , Ishikawa , Japan
| | - Samantha A Martin
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Matthew A Smith
- c Department of Pathology and Laboratory Medicine , Allegheny Health Network , Pittsburgh , PA , USA
| | - D Aaron Guel
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Jenna Kolano
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Yoshihiro Komatsu
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Fahim Habib
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Christopher Lai
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Kevi Christopher
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Ronan J Kelly
- d Department of Oncology, Sidney Kimmel Comprehensive Cancer Center , Johns Hopkins Hospital , Baltimore , MD , USA
| | - Ali H Zaidi
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Blair A Jobe
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| |
Collapse
|
11
|
Kosovec JE, Zaidi AH, Omstead AN, Matsui D, Biedka MJ, Cox EJ, Campbell PT, Biederman RW, Kelly RJ, Jobe BA. CDK4/6 dual inhibitor abemaciclib demonstrates compelling preclinical activity against esophageal adenocarcinoma: a novel therapeutic option for a deadly disease. Oncotarget 2017; 8:100421-100432. [PMID: 29245989 PMCID: PMC5725031 DOI: 10.18632/oncotarget.22244] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/16/2017] [Indexed: 12/21/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is a deadly disease with limited therapeutic options. In the present study, we determined the preclinical efficacy of CDK4/6 inhibitor abemaciclib for treatment of EAC. In vitro, apoptosis, proliferation, and pathway regulation were evaluated in OE19, OE33, and FLO1 EAC cell lines. In vivo, esophagojejunostomy was performed on rats to induce EAC. At 36 weeks post-surgery, MRI and endoscopic biopsy established baseline tumor volume and molecular correlates, respectively. Next, the study animals were randomized to 26mg/kg intraperitoneal abemaciclib treatment or vehicle control for 28 days. Pre and post treatment MRIs, histopathology, and qRT-PCR were utilized to determine response. Our results demonstrated treatment with abemaciclib lead to increased apoptosis, and decreased proliferation in OE19 (p=0.185), OE33 (p=0.048), and FLO1 (p=0.043) with anticipated downstream molecular inhibition. In vivo, 78.9% of treatment animals demonstrated >20% tumor volume decrease (placebo 0%). Mean tumor volume changed in the treatment arm by -65.5% (placebo +133.5%) (p<0.01), and prevalence changed by -37.5% (placebo +16.7%) (p<0.01). Pre vs post treatment qRT-PCR demonstrated significant inhibition of all downstream molecular correlates. Overall our findings suggest potent antitumor efficacy of abemaciclib against EAC with evident molecular pathway inhibition and reasonable safety, establishing the rationale for future clinical development.
Collapse
Affiliation(s)
- Juliann E. Kosovec
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ali H. Zaidi
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ashten N. Omstead
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Daisuke Matsui
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Mark J. Biedka
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Erin J. Cox
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Patrick T. Campbell
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | | | - Ronan J. Kelly
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Blair A. Jobe
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Matsui D, Omstead AN, Kosovec JE, Komatsu Y, Lloyd EJ, Raphael H, Kelly RJ, Zaidi AH, Jobe BA. High yield reproducible rat model recapitulating human Barrett’s carcinogenesis. World J Gastroenterol 2017; 23:6077-6087. [PMID: 28970723 PMCID: PMC5597499 DOI: 10.3748/wjg.v23.i33.6077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/23/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To efficiently replicate the biology and pathogenesis of human esophageal adenocarcinoma (EAC) using the modified Levrat model of end-to-side esophagojejunostomy.
METHODS End-to-side esophagojejunostomy was performed on rats to induce gastroduodenoesophageal reflux to develop EAC. Animals were randomly selected and serially euthanized at 10 (n = 6), 17 (n = 8), 24 (n = 9), 31 (n = 6), 38 (n = 6), and 40 (n = 6) wk postoperatively. The esophagi were harvested for downstream histopathology and gene expression. Histological evaluation was completed to determine respective rates of carcinogenic development. Quantitative reverse transcription-polymerase chain reaction was performed to determine gene expression levels of MUC2, CK19, and CK20, and results were compared to determine significant differences throughout disease progression stages.
RESULTS The overall study mortality was 15%. Causes of mortality included anastomotic leak, gastrointestinal hemorrhage, stomach ulcer perforation, respiratory infection secondary to aspiration, and obstruction due to tumor or late anastomotic stricture. 10 wk following surgery, 100% of animals presented with esophagitis. Barrett’s esophagus (BE) was first observed at 10 wk, and was present in 100% of animals by 17 wk. Dysplasia was confirmed in 87.5% of animals at 17 wk, and increased to 100% by 31 wk. EAC was first observed in 44.4% of animals at 24 wk and increased to 100% by 40 wk. In addition, two animals at 38-40 wk post-surgery had confirmed macro-metastases in the lung/liver and small intestine, respectively. MUC2 gene expression was progressively down-regulated from BE to dysplasia to EAC. Both CK19 and CK20 gene expression significantly increased in a stepwise manner from esophagitis to EAC.
CONCLUSION Esophagojejunostomy was successfully replicated in rats with low mortality and a high tumor burden, which may facilitate broader adoption to study EAC development, progression, and therapeutics.
Collapse
Affiliation(s)
- Daisuke Matsui
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Ishikawa 920-1192, Japan
| | - Ashten N Omstead
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
| | - Juliann E Kosovec
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
| | - Yoshihiro Komatsu
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
| | - Emily J Lloyd
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
| | - Hailey Raphael
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
| | - Ronan J Kelly
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD 21231, United States
| | - Ali H Zaidi
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
| | - Blair A Jobe
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA 15224, United States
| |
Collapse
|