Hyperthermic Intraperitoneal Chemotherapy-Induced Molecular Changes in Humans Validate Preclinical Data in Ovarian Cancer

Thanh H Dellinger, Ernest S Han, Mustafa Raoof, Byrne Lee, Xiwei Wu, Hyejin Cho, Ting-Fang He, Peter Lee, Marianne Razavi, Winnie S Liang, Daniel Schmolze, Saul J Priceman, Stephen Lee, Wei-Chien Lin, Jeff F Lin, Mehdi Kebria, Amy Hakim, Nora Ruel, Daphne B Stewart, Edward W Wang, Benjamin I Paz, Mark T Wakabayashi, Mihaela C Cristea, Lorna Rodriguez-Rodriguez, Thanh H Dellinger, Ernest S Han, Mustafa Raoof, Byrne Lee, Xiwei Wu, Hyejin Cho, Ting-Fang He, Peter Lee, Marianne Razavi, Winnie S Liang, Daniel Schmolze, Saul J Priceman, Stephen Lee, Wei-Chien Lin, Jeff F Lin, Mehdi Kebria, Amy Hakim, Nora Ruel, Daphne B Stewart, Edward W Wang, Benjamin I Paz, Mark T Wakabayashi, Mihaela C Cristea, Lorna Rodriguez-Rodriguez

Abstract

Purpose: Hyperthermic intraperitoneal chemotherapy (HIPEC) confers a survival benefit in epithelial ovarian cancer (EOC) and in preclinical models. However, the molecular changes induced by HIPEC have not been corroborated in humans.

Patients and methods: A feasibility trial evaluated clinical and safety outcomes of HIPEC with cisplatin during optimal cytoreductive surgery (CRS) in patients with EOC diagnosed with stage III, IV, or recurrent EOC. Pre- and post-HIPEC biopsies were comprehensively profiled with genomic and transcriptomic sequencing to identify mutational and RNAseq signatures correlating with response; the tumor microenvironment was profiled to identify potential immune biomarkers; and transcriptional signatures of tumors and normal samples before and after HIPEC were compared to investigate HIPEC-induced acute transcriptional changes.

Results: Thirty-five patients had HIPEC at the time of optimal CRS; all patients had optimal CRS. The median progression-free survival (PFS) was 24.7 months for primary patients and 22.4 for recurrent patients. There were no grade 4 or 5 adverse events. Anemia was the most common grade 3 adverse event (43%). Hierarchical cluster analyses identified distinct transcriptomic signatures of good versus poor responders to HIPEC correlating with a PFS of 29.9 versus 7.3 months, respectively. Among good responders, significant HIPEC-induced molecular changes included immune pathway upregulation and DNA repair pathway downregulation. Within cancer islands, % programmed cell death protein 1 expression in CD8+ T cells significantly increased after HIPEC. An exceptional responder (PFS 58 months) demonstrated the highest programmed cell death protein 1 increase. Heat shock proteins comprised the top differentially upregulated genes in HIPEC-treated tumors.

Conclusion: Distinct transcriptomic signatures identify responders to HIPEC, and preclinical model findings are confirmed for the first time in a human cohort.

Trial registration: ClinicalTrials.gov NCT01970722.

Conflict of interest statement

Ernest S. HanResearch Funding: Vergent Bioscience (Inst) Mustafa RaoofOpen Payments Link: https://openpaymentsdata.cms.gov/physician/3634542 Saul J. PricemanStock and Other Ownership Interests: Imugene Ltd, Adicet BioConsulting or Advisory Role: Imugene Ltd, Adicet Bio, MustangBio, BayerResearch Funding: Imugene Ltd, Carisma TherapeuticsPatents, Royalties, Other Intellectual Property: CAR Therapeutic, Oncolytic Virus Therapeutic Jeff F. LinHonoraria: C-SATSConsulting or Advisory Role: Aspira Women's HealthTravel, Accommodations, Expenses: Intuitive Surgical, GlaxoSmithKline Mehdi KebriaHonoraria: GlaxoSmithKlineSpeakers' Bureau: GlaxoSmithKline Nora RuelConsulting or Advisory Role: EPHOS Bioscience Benjamin I. PazEmployment: City of HopeStock and Other Ownership Interests: Bio-Path Holdings Inc Mark T. WakabayashiEmployment: RegeneronStock and Other Ownership Interests: Regeneron Mihaela C. CristeaHonoraria: AstraZeneca, AbbVieConsulting or Advisory Role: AstraZeneca, AbbVieSpeakers' Bureau: AstraZenecaNo other potential conflicts of interest were reported.

Figures

FIG 1.
FIG 1.
Study schema: (A) study flow and (B) flow chart of data processing and analysis. HIPEC, hyperthermic intraperitoneal chemotherapy; PFS, progression-free survival.
FIG 2.
FIG 2.
Patient outcomes (survival and safety): (A) Swimmer plot showing disease status and outcomes for all patients with a follow-up of 72 months. (B) Kaplan-Meier survival curve depicting PFS and OS for all patients. (C) AEs—treatment-related toxicity. Bars represent on-study and follow-up period. AE, adverse event; NR, not reached; OS, overall survival; PFS, progression-free survival.
FIG 3.
FIG 3.
Outcome-related gene and mutational signatures of HIPEC responders. (A) Hierarchical clustered analysis of significantly changed genes in tumor samples of 15 patients. Responders were categorized into good responders (PFS > 12 months) and poor responders (PFS

FIG 4.

Tumor microenvironment changes induced by…

FIG 4.

Tumor microenvironment changes induced by HIPEC. Multiplex immunofluorescence estimation of tumor-infiltrating immune subsets…

FIG 4.
Tumor microenvironment changes induced by HIPEC. Multiplex immunofluorescence estimation of tumor-infiltrating immune subsets and PD-1 expression in matched pre- and post HIPEC tumors. (A) Pre- and post-HIPEC tumors were stained with TIL markers (CD3, CD8, FOXP3), PD-1, for the following phenotypes: CD4+ conventional T cells (CD3+ CD8– FOXP3– cells), CD4+ regulatory T cells (CD3+ CD8– FOXP3+), and CD8+ T cells (CD3+ CD8+). (B) Cell density of CD8+ T cells, CD4+ conventional T cells, and CD4+ regulatory T cells do not change with HIPEC treatment. (C) Evaluation of PD-1 expression in CD8+ T cells. PD-1 expression rises in CD8+ T cells within cancer islands (cancer), but not in stroma. (D) Representative immunofluorescence demonstrating increased PD-1 staining of CD8+ T cells within cancer islands after HIPEC while stromal PD-1 expression remained stable after HIPEC. (E) CK was used as a marker for cancer islands to delineate it from stroma. (F) PD-1 expression changes in individual patients before and after HIPEC in CD8+ T cells (stroma). The patient with the highest rise in %PD-1 was patient 1, an exceptional responder with PFS of 5 years and demonstrated the largest PD-1 increase. The only patient with decreased PD-1 expression was patient 8, who was the only patient with clear cell histology and had a poor survival (PFS 24 months) and poor (PFS

FIG 5.

Gene and pathway changes of…

FIG 5.

Gene and pathway changes of pre- and post-HIPEC in metastatic tumor and normal…

FIG 5.
Gene and pathway changes of pre- and post-HIPEC in metastatic tumor and normal patient samples. (A and B) Hierarchical clustered analysis of (A) differentially expressed genes in metastatic tumors and (B) normal samples. Cluster 1 was enriched for pre-HIPEC; cluster 2 was enriched with post-HIPEC. (C) Volcano plot displaying gene expression post-HIPEC by log2-fold change (x-axis) and minus log10P value (y-axis) in tumor samples. Significantly upregulated genes of ≥ 2-fold expression are in red while statistically significantly downregulated genes of ≥ 2-fold expression are in green. The top five differentially expressed genes with fold change (post- v pre-HIPEC) and P value shown for tumor samples are shown. (D) Volcano plot displaying gene expression post-HIPEC by log2-fold change (x-axis) and minus log10P value (y-axis) in normal samples. (E and F) Kegg pathway gene set enrichment analysis of (E) HIPEC metastatic tumors and (F) normal samples demonstrating the top significantly upregulated and downregulated pathways (FDR < 0.05). Several immune-related pathways are upregulated, and DNA replication pathways are downregulated in tumor samples. Normal HIPEC samples demonstrate significant downregulation of metabolic pathways. FC, fold change; FDR, false discovery rate; HIPEC, hyperthermic intraperitoneal chemotherapy; NES, normalized enrichment score; q. val., q value; TCA, tricarboxylic acid.
Similar articles
Cited by
References
    1. de Bree E, Tsiftsis DD: Experimental and pharmacokinetic studies in intraperitoneal chemotherapy: From laboratory bench to bedside. Recent Results Cancer Res 169:53-73, 2007 - PubMed
    1. Armstrong DK, Bundy B, Wenzel L, et al. : Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med 354:34-43, 2006 - PubMed
    1. Mackay HJ, Kohn EC: Intraperitoneal chemotherapy: Hot, timely, and relevant? Cancer 126:5206-5209, 2020 - PubMed
    1. Dellinger TH, Han ES: State of the Science: The role of HIPEC in the treatment of ovarian cancer. Gynecol Oncol 160:364-368, 2021 - PubMed
    1. van Driel WJ, Koole SN, Sikorska K, et al. : Hyperthermic intraperitoneal chemotherapy in ovarian cancer. N Engl J Med 378:230-240, 2018 - PubMed
Show all 31 references
Publication types
MeSH terms
Associated data
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
FIG 4.
FIG 4.
Tumor microenvironment changes induced by HIPEC. Multiplex immunofluorescence estimation of tumor-infiltrating immune subsets and PD-1 expression in matched pre- and post HIPEC tumors. (A) Pre- and post-HIPEC tumors were stained with TIL markers (CD3, CD8, FOXP3), PD-1, for the following phenotypes: CD4+ conventional T cells (CD3+ CD8– FOXP3– cells), CD4+ regulatory T cells (CD3+ CD8– FOXP3+), and CD8+ T cells (CD3+ CD8+). (B) Cell density of CD8+ T cells, CD4+ conventional T cells, and CD4+ regulatory T cells do not change with HIPEC treatment. (C) Evaluation of PD-1 expression in CD8+ T cells. PD-1 expression rises in CD8+ T cells within cancer islands (cancer), but not in stroma. (D) Representative immunofluorescence demonstrating increased PD-1 staining of CD8+ T cells within cancer islands after HIPEC while stromal PD-1 expression remained stable after HIPEC. (E) CK was used as a marker for cancer islands to delineate it from stroma. (F) PD-1 expression changes in individual patients before and after HIPEC in CD8+ T cells (stroma). The patient with the highest rise in %PD-1 was patient 1, an exceptional responder with PFS of 5 years and demonstrated the largest PD-1 increase. The only patient with decreased PD-1 expression was patient 8, who was the only patient with clear cell histology and had a poor survival (PFS 24 months) and poor (PFS

FIG 5.

Gene and pathway changes of…

FIG 5.

Gene and pathway changes of pre- and post-HIPEC in metastatic tumor and normal…

FIG 5.
Gene and pathway changes of pre- and post-HIPEC in metastatic tumor and normal patient samples. (A and B) Hierarchical clustered analysis of (A) differentially expressed genes in metastatic tumors and (B) normal samples. Cluster 1 was enriched for pre-HIPEC; cluster 2 was enriched with post-HIPEC. (C) Volcano plot displaying gene expression post-HIPEC by log2-fold change (x-axis) and minus log10P value (y-axis) in tumor samples. Significantly upregulated genes of ≥ 2-fold expression are in red while statistically significantly downregulated genes of ≥ 2-fold expression are in green. The top five differentially expressed genes with fold change (post- v pre-HIPEC) and P value shown for tumor samples are shown. (D) Volcano plot displaying gene expression post-HIPEC by log2-fold change (x-axis) and minus log10P value (y-axis) in normal samples. (E and F) Kegg pathway gene set enrichment analysis of (E) HIPEC metastatic tumors and (F) normal samples demonstrating the top significantly upregulated and downregulated pathways (FDR < 0.05). Several immune-related pathways are upregulated, and DNA replication pathways are downregulated in tumor samples. Normal HIPEC samples demonstrate significant downregulation of metabolic pathways. FC, fold change; FDR, false discovery rate; HIPEC, hyperthermic intraperitoneal chemotherapy; NES, normalized enrichment score; q. val., q value; TCA, tricarboxylic acid.
FIG 5.
FIG 5.
Gene and pathway changes of pre- and post-HIPEC in metastatic tumor and normal patient samples. (A and B) Hierarchical clustered analysis of (A) differentially expressed genes in metastatic tumors and (B) normal samples. Cluster 1 was enriched for pre-HIPEC; cluster 2 was enriched with post-HIPEC. (C) Volcano plot displaying gene expression post-HIPEC by log2-fold change (x-axis) and minus log10P value (y-axis) in tumor samples. Significantly upregulated genes of ≥ 2-fold expression are in red while statistically significantly downregulated genes of ≥ 2-fold expression are in green. The top five differentially expressed genes with fold change (post- v pre-HIPEC) and P value shown for tumor samples are shown. (D) Volcano plot displaying gene expression post-HIPEC by log2-fold change (x-axis) and minus log10P value (y-axis) in normal samples. (E and F) Kegg pathway gene set enrichment analysis of (E) HIPEC metastatic tumors and (F) normal samples demonstrating the top significantly upregulated and downregulated pathways (FDR < 0.05). Several immune-related pathways are upregulated, and DNA replication pathways are downregulated in tumor samples. Normal HIPEC samples demonstrate significant downregulation of metabolic pathways. FC, fold change; FDR, false discovery rate; HIPEC, hyperthermic intraperitoneal chemotherapy; NES, normalized enrichment score; q. val., q value; TCA, tricarboxylic acid.

References

    1. de Bree E, Tsiftsis DD: Experimental and pharmacokinetic studies in intraperitoneal chemotherapy: From laboratory bench to bedside. Recent Results Cancer Res 169:53-73, 2007
    1. Armstrong DK, Bundy B, Wenzel L, et al. : Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med 354:34-43, 2006
    1. Mackay HJ, Kohn EC: Intraperitoneal chemotherapy: Hot, timely, and relevant? Cancer 126:5206-5209, 2020
    1. Dellinger TH, Han ES: State of the Science: The role of HIPEC in the treatment of ovarian cancer. Gynecol Oncol 160:364-368, 2021
    1. van Driel WJ, Koole SN, Sikorska K, et al. : Hyperthermic intraperitoneal chemotherapy in ovarian cancer. N Engl J Med 378:230-240, 2018
    1. Lavoue V, Bakrin N, Bolze PA, et al. : Saved by the evidence: Hyperthermic intraperitoneal chemotherapy still has a role to play in ovarian cancer. Eur J Surg Oncol 45:1757-1759, 2019
    1. Vermorken JB, van Dam P, Brand A: HIPEC in advanced epithelial ovarian cancer: Why is there controversy? Curr Opin Oncol 32:451-458, 2020
    1. Fotopoulou C, Sehouli J, Mahner S, et al. : HIPEC: HOPE or HYPE in the fight against advanced ovarian cancer? Ann Oncol 29:1610-1613, 2018
    1. Vergote I, Harter P, Chiva L: Hyperthermic intraperitoneal chemotherapy does not improve survival in advanced ovarian cancer. Cancer 125:4594-4597, 2019. (suppl 24)
    1. Vergote I, Harter P, Chiva L: Is there a role for intraperitoneal chemotherapy, including HIPEC, in the management of ovarian cancer? J Clin Oncol 37:2420-2423, 2019
    1. Chen T, Guo J, Han C, et al. : Heat shock protein 70, released from heat-stressed tumor cells, initiates antitumor immunity by inducing tumor cell chemokine production and activating dendritic cells via TLR4 pathway. J Immunol 182:1449-1459, 2009
    1. Zunino B, Rubio-Patino C, Villa E, et al. : Hyperthermic intraperitoneal chemotherapy leads to an anticancer immune response via exposure of cell surface heat shock protein 90. Oncogene 35:261-268, 2016
    1. Ahmed K, Tabuchi Y, Kondo T: Hyperthermia: An effective strategy to induce apoptosis in cancer cells. Apoptosis 20:1411-1419, 2015
    1. Mantso T, Goussetis G, Franco R, et al. : Effects of hyperthermia as a mitigation strategy in DNA damage-based cancer therapies. Semin Cancer Biol 37-38: 96-105, 2016
    1. Lesnock JL, Darcy KM, Tian C, et al. : BRCA1 expression and improved survival in ovarian cancer patients treated with intraperitoneal cisplatin and paclitaxel: A Gynecologic Oncology Group study. Br J Cancer 108:1231-1237, 2013
    1. Naumann RW, Morris JC, Tait DL, et al. : Patients with BRCA mutations have superior outcomes after intraperitoneal chemotherapy in optimally resected high grade ovarian cancer. Gynecol Oncol 151:477-480, 2018
    1. Dellinger TH, Smith DD, Ballard E, et al. : HIPEC treatment in advanced and recurrent ovarian cancer: A meta-analysis of observational studies. Eur J Gynaecol Oncol 39:353-360, 2018
    1. Sugarbaker PH: Intraperitoneal chemotherapy and cytoreductive surgery for the prevention and treatment of peritoneal carcinomatosis and sarcomatosis. Semin Surg Oncol 14:254-261, 1998
    1. Robinson MD, McCarthy DJ, Smyth GK: edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26:139-140, 2010
    1. Subramanian A, Tamayo P, Mootha VK, et al. : Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 102:15545-15550, 2005
    1. de Hoon MJ, Imoto S, Nolan J, et al. : Open source clustering software. Bioinformatics 20:1453-1454, 2004
    1. Saldanha AJ: Java Treeview—Extensible visualization of microarray data. Bioinformatics 20:3246-3248, 2004
    1. Thakur B, Ray P: Cisplatin triggers cancer stem cell enrichment in platinum-resistant cells through NF-kappaB-TNFalpha-PIK3CA loop. J Exp Clin Cancer Res 36:164, 2017
    1. Roth L, Eshmuminov D, Laminger F, et al. : Systemic inflammatory response after hyperthermic intraperitoneal chemotherapy (HIPEC): The perfusion protocol matters! Eur J Surg Oncol 45:1734-1739, 2019
    1. Sharpe AH, Pauken KE: The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18:153-167, 2018
    1. Calderwood SK, Khaleque MA, Sawyer DB, et al. : Heat shock proteins in cancer: Chaperones of tumorigenesis. Trends Biochem Sci 31:164-172, 2006
    1. Skitzki JJ, Repasky EA, Evans SS: Hyperthermia as an immunotherapy strategy for cancer. Curr Opin Investig Drugs 10:550-558, 2009
    1. Dahl O: Interaction of heat and drugs in vitro and in vivo, in Seegenschmiedt M, Fessenden P, Vernon C (eds): Thermoradiotherapy and Thermochemotherapy. Berlin, Springer, 1995, pp 103-121
    1. Warters RL, Henle KJ: DNA degradation in Chinese hamster ovary cells after exposure to hyperthermia. Cancer Res 42:4427-4432, 1982
    1. Hurwitz M, Stauffer P: Hyperthermia, radiation and chemotherapy: The role of heat in multidisciplinary cancer care. Semin Oncol 41:714-729, 2014
    1. Zivanovic O, Chi D, Zhou Q, et al. : A randomized phase II trial of secondary cytoreductive surgery (SCS) +/- carboplatin hyperthermic intraperitoneal chemotherapy (HIPEC) in patients (pts) with recurrent platinum-sensitive ovarian cancer (EOC). J Clin Oncol 38, 2020. (15_suppl; abstr 6016)

Source: PubMed

3
Abonneren