Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Clinical Trial
. 2024 Feb 20;5(2):101393.
doi: 10.1016/j.xcrm.2024.101393. Epub 2024 Jan 26.

Immunomodulatory effects and improved outcomes with cisplatin- versus carboplatin-based chemotherapy plus atezolizumab in urothelial cancer

Affiliations
Clinical Trial

Immunomodulatory effects and improved outcomes with cisplatin- versus carboplatin-based chemotherapy plus atezolizumab in urothelial cancer

Matthew D Galsky et al. Cell Rep Med. .

Abstract

In metastatic urothelial cancer (mUC), cisplatin versus carboplatin leads to durable disease control in a subset of patients. The IMvigor130 trial reveals more favorable effects with atezolizumab combined with gemcitabine and cisplatin (GemCis) versus gemcitabine and carboplatin (GemCarbo). This study investigates the immunomodulatory effects of cisplatin as a potential explanation for these observations. Our findings indicate that improved outcomes with GemCis versus GemCarbo are primarily observed in patients with pretreatment tumors exhibiting features of restrained adaptive immunity. In addition, GemCis versus GemCarbo ± atezolizumab induces transcriptional changes in circulating immune cells, including upregulation of antigen presentation and T cell activation programs. In vitro experiments demonstrate that cisplatin, compared with carboplatin, exerts direct immunomodulatory effects on cancer cells, promoting dendritic cell activation and antigen-specific T cell killing. These results underscore the key role of immune modulation in cisplatin's efficacy in mUC and highlight the importance of specific chemotherapy backbones in immunotherapy combination regimens.

Trial registration: ClinicalTrials.gov NCT02807636.

Keywords: PD-1 blockade; PD-L1 blockade; bladder cancer; carboplatin; chemotherapy; cisplatin; immune checkpoint blockade; immunogenic cell death; single cell RNA sequencing; urothelial cancer.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests M.D.G. has received grants or contracts from Bristol Myers Squibb, Novartis, Dendreon, AstraZeneca, and Merck; and has received consulting fees from Bristol Myers Squibb, Merck, Genentech Inc., AstraZeneca, Pfizer, EMD Serono, Seagen, Janssen, Numab, Dragonfly, GlaxoSmithKline, Basilea, UroGen, RapptaTherapeutics, Alligator, Silverback, Fujifilm, and Curis. X.G., D.R., A.S.R., H.M.S., R.B., K.Y., E.V., R.H., C.-J.H., Y.L., D.V., P.W., I.M., S.S., R.J., and S.M. are employees of Genentech Inc., and hold stocks or stock options in Roche/Genentech Inc. H.L. is an employee of Hoffmann-La Roche Ltd, Canada. L.W., J.Z., and H.Y. are employees of Sema4. E.H. was employed at Immunai during study conduct. R.H.H. and E. Kiner are employees of Immunai. A.B. has served as a consultant or advisor to Roche, Bristol Myers Squibb, MSD, and Pfizer; has received honoraria from Bristol Myers Squibb, MSD, and Pfizer; and has received educational and research grants from Pfizer, Pierre-Fabre, and Bristol Myers Squibb. M.D.S. has received consulting fees from AAA, Accord, Amgen, Astellas, AstraZeneca, Basilea, Bayer, Bioclin, Bristol Myers Squibb, Eisai, Ferring, Immunomedics, Ipsen, Janssen, MSD, Merck, Novartis, Pfizer, Pierre Fabre Oncology, Roche, Sandoz, Sanofi, and Seagen; has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing, or educational events from AAA, Accord, Amgen, Astellas, AstraZeneca, Basilea, Bayer, Bioclin, Bristol Myers Squibb, Eisai, Ferring, Immunomedics, Ipsen, Janssen, MSD, Merck, Novartis, Pfizer, Pierre Fabre Oncology, Roche, Sandoz, Sanofi, and Seagen; has received support for attending meetings and/or travel from AAA, Accord, Amgen, Astellas, AstraZeneca, Basilea, Bayer, Bioclin, Bristol Myers Squibb, Eisai, Ferring, Immunomedics, Ipsen, Janssen, MSD, Merck, Novartis, Pfizer, Pierre Fabre Oncology, Roche, Sandoz, Sanofi, and Seagen; has participated in a Data Safety Monitoring Board or Advisory Board for Roche, Orion, and CR-UK; and has other financial or non-financial interests by having worked on the ESMO guidelines on bladder cancer and the German S3 Leitlinie Blasenkarzinom. I.D.D. has participated in a Data Safety Monitoring Board or Advisory Board for Roche/Genentech Inc. (WO30070 [IMvigor130] international steering committee; unpaid) and Merck/Pfizer (APAC GU Advisory Board; unpaid: honoraria are invoiced by and paid directly to ANZUP Cancer Trials Group); and has a leadership or fiduciary role in the ANZUP Cancer Trials Group (as a Director and Board Chair; unpaid). J.A.A. has received third-party service as collaboration in the selection, and management of administrative requirements for the participation of SOGUG centers in the study from SOGUG during the conduct of the study; speaking or consulting fees from Bristol Myers Squibb, MSD, Roche, Astellas, Janssen Cilag, Pfizer, Novartis, and Bayer outside the submitted work; travel support from Bristol Myers Squibb, MSD, Roche, and Janssen Cilag outside the submitted work; research funding (from SOGUG) outside the submitted work from Bristol Myers Squibb, Novartis, and Pierre Fabre; and has participated in industry-sponsored clinical trials for Bristol Myers Squibb, MSD, Roche, Astellas, Janssen Cilag, Pfizer, and Novartis outside the submitted work. E. Kikuchi has received grants or contracts (through his institution) from Takeda, Nippon Kayaku, and Taiho; has received consulting fees from Chugai, Nippon Kayaku, MSD, Takeda, Pfizer, Astellas, Bristol Myers Squibb, and Yansen; has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing, or educational events from Takeda, Nippon Kayaku, and Taiho; and has received consulting fees from Chugai, Nippon Kayaku, MSD, Takeda, Taiho, AstraZeneca, Pfizer, Astellas, Bristol Myers Squibb, and Yansen. S.B. is an employee of Roche Products Ltd, UK, holds stocks or stock options in Roche, and has other financial or non-financial interests in Roche. C.L. is an employee of Roche Products Ltd, UK, holds stocks or stock options in Roche, and has other financial or non-financial interests in Roche. P.C.B. has participated in advisory boards for Janssen, Merck, Roche/Genentech Inc., Bristol Myers Squibb, Urogen, EMD Serono, Bayer, Astellas, AbbVie, AstraZeneca, Ferring, H3-Biomedicine, Sanofi, Pfizer, Prokarium, Protara Therapeutics, Stimit, and Verity; has received payment or honoraria as a speaker from Janssen, Bayer, Ferring, H3-Biomedicine, and Pfizer; has participated in clinical trials for Roche/Genentech Inc., Bristol Myers Squibb, and AstraZeneca; and has received non-financial support (scientific collaboration on bladder cancer genomics/transcriptomics) from Decipher Biosciences/Veracyte. E.G. has received grants or contracts from Astellas, AstraZeneca, Ipsen, Merck KGaA, and Pfizer; has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing, or educational events from Astellas, AstraZeneca, Bayer, Bristol Myers Squibb, Eisai, Esteve, Ipsen, Janssen, Lilly, Merck KGaA, MSD, Pfizer, Raffo, and Roche; and has received support for attending meetings and/or travel from Roche, AstraZeneca, Bristol Myers Squibb, Pfizer, and Merck KGaA.

Figures

None
Graphical abstract
Figure 1
Figure 1
Survival outcomes with GemCis but not with GemCarbo are dependent on pretreatment tumor PD-L1 expression in an exploratory analysis of the IMvigor130 study (A) Study design of phase 3 IMvigor130. Patients in arms A and C received atezolizumab and placebo, respectively, in combination with the investigators’ choice of platinum drugs (cisplatin versus carboplatin) and gemcitabine. Arm A, n = 451; arm B, n = 362; arm C, n = 400. (B) Forest plot showing overall survival in patients in arms A and C by use of GemCis versus GemCarbo. Hazard ratios, 95% confidence intervals, and p values were calculated using a univariate Cox model. The diamonds represent the hazard ratios, and the horizontal bars their 95% confidence intervals. (C) Kaplan-Meier curves showing overall survival in patients in arm C stratified by PD-L1 status (IC2/3 versus IC0/1) and use of GemCis (left) or GemCarbo (right). (D) Kaplan-Meier curves showing overall survival in patients in arm A stratified by PD-L1 status (IC2/3 versus IC0/1) and use of GemCis plus atezolizumab (left) or GemCarbo plus atezolizumab (right). In (C) and (D), p values were estimated using the log rank test. Hazard ratios and 95% confidence intervals were calculated using a univariate Cox model. (E) Kaplan-Meier curves showing overall survival in patients in arm C stratified by actual receipt of GemCis or GemCarbo and tumor PD-L1 status (IC2/3 versus IC0/1). Patients classified as “cisplatin-eligible” according to standard criteria were included. p values were estimated using the log rank test. See also Figures S1 and S2 and Tables S1 and S2.
Figure 2
Figure 2
PD-L1 IC2/3 versus IC0/1 tumors are enriched in immune-related gene signatures (A) Heatmap detailing immune-related gene signatures based on bulk RNA sequencing of pretreatment archival tumor specimens from the IMvigor130 study according to PD-L1 status (IC2/3 and IC0/1) and treatment arm. (B) Volcano plot detailing differentially expressed genes based on bulk RNA sequencing of pretreatment archival tumor specimens from the IMvigor130 study according to PD-L1 status (IC2/3 versus IC0/1). Differential expression analysis was conducted with limma-based statistical methods and the Benjamini-Hochberg correction. (C) Gene set enrichment analysis based on bulk RNA sequencing of pretreatment archival tumor specimens from the IMvigor130 study according to PD-L1 status (IC2/3 versus IC0/1) and treatment arm or specific platinum drug received. The hue represents the false discovery rate (FDR) significance derived from the fgsea package. Black asterisks represent FDR < 0.05.
Figure 3
Figure 3
GemCis ± atezolizumab versus GemCarbo ± atezolizumab induces proinflammatory transcriptional programs across multiple immune cell subsets in PBMCs (A) Single-cell CITE-seq experimental design and analysis workflow. (B) Uniform Manifold Approximation and Projection (UMAP) visualization of single cells captured, colored by major cell types (n = 865,922) (left), and heatmap showing scaled expression of the canonical marker genes across cell types (right). (C and D) Heatmaps showing pathway enrichment on-treatment (C3D1) versus at baseline (C1D1) across multiple immune cell types. Treatments received were GemCis (left, n = 17 pairs) or GemCarbo (right, n = 16 pairs) in arm C (C) and atezolizumab (n = 33 pairs) in arm B (D). (E) Comparison of MHC class gene expression on-treatment (C3D1) versus at baseline (C1D1) in monocytes collected from patients receiving GemCis or GemCarbo in arm C. (F) Heatmaps showing pathway enrichment on-treatment (C3D1) versus at baseline (C1D1) across multiple immune cell types. Treatments received were GemCis + atezolizumab (left, n = 14 pairs) or GemCarbo + atezolizumab (right, n = 24 pairs) in arm A. In (C), (D), and (F), red indicates enrichment in on-treatment samples and blue indicates enrichment at baseline. The hue represents the false discovery rate (FDR) significance derived from the fgsea package. Black asterisks represent FDR < 0.05. See also Figures S3 and S4.
Figure 4
Figure 4
Atezolizumab added to GemCis versus GemCarbo leads to distinct transcriptional states of circulating immune cells including changes reflective of T cell activation states (A) UMAP visualization of total T and NK cells captured, colored by major cell types (n = 707,614). (B) UMAP visualization of total CD8 T cells captured, colored by different cell types (n = 171,729) (left), and heatmap showing scaled expression of the top 10 cell markers ranked by fold change in each cell type (right). Black asterisks indicate cell types not included in subsequent analysis due to low cell numbers. (C) Heatmaps showing pathway enrichment on-treatment (C3D1) versus at baseline (C1D1) across T and NK cell types. Treatments received were GemCis (first panel, n = 17 pairs) or GemCarbo (second panel, n = 16 pairs) in arm C and GemCis + atezolizumab (third panel, n = 14 pairs) or GemCarbo + atezolizumab (fourth panel, n = 24 pairs) in arm A. (D) Heatmaps showing pathway enrichment with GemCis on-treatment (C3D1) versus baseline (C1D1) across T and NK cell types in responders (left) and nonresponders (right). In (C) and (D), red indicates enrichment in on-treatment samples and blue indicates enrichment at baseline. The hue represents the false discovery rate (FDR) significance derived from the fgsea package. Black asterisks represent FDR < 0.05.
Figure 5
Figure 5
The direct effects of cisplatin versus carboplatin on downstream immune-related programs in cancer cells are mediated by the DNA damage transducer ATR (A) Enrichment of Hallmark gene sets with cisplatin (red) versus carboplatin (gray) treatment in the 5637 human bladder cancer cell line (top), RT112 human bladder cancer cell line (middle), and MC38 murine colon cancer cell line (bottom). The cisplatin and carboplatin concentrations used were 5 and 35 μM, respectively, and cells were collected 24 h after treatment. Only pathways that were significantly changed with cisplatin versus carboplatin (false discovery rate [FDR] < 0.05) are shown. n = 3 per treatment group for the 5637 and RT112 cell lines; n = 2 per treatment group for the MC38 cell line. (B) Protein expression of PD-L1 as determined by the median fluorescence intensity in the three cell lines treated with increasing concentrations of cisplatin (red) or carboplatin (black) for 24 h. Data depict the aggregate of three independent experiments (mean ± SEM). (C) Representative immunoblots showing p-ATM S1981, p-ATR T1989, p-Chk1 S317, p-Chk2 T68, and GAPDH levels in lysates of the 5637 cell line treated with increasing concentrations of cisplatin or carboplatin for 6 h. One representative experiment out of four independent experiments is shown. (D) Protein expression of PD-L1 as determined by median fluorescence intensity in the 5637 cell line treated with increasing concentrations of cisplatin or carboplatin for 24 h, in the presence or absence of an ATM inhibitor (KU-55933, 1 μM) or ATR inhibitor (VE-821, 1 μM). Data depict the aggregate of three independent experiments (mean ± SEM). (E and F) Bulk RNA-seq analysis of 5637 cells treated with 5 μM cisplatin or 35 μM carboplatin, in the absence (gold) or presence of 1 μM ATR inhibitor (navy blue) for 24 h. (E) Enrichment of Hallmark gene sets. n = 4 per treatment group. (F) Heatmap displaying the scaled transcriptional expression of genes involved in immune-related transcriptional programs. Heatmap shows an average score of n = 4 individual samples per treatment group. See also Figures S5–S8.
Figure 6
Figure 6
Co-culture of cisplatin- versus carboplatin-pretreated MC38 cancer cells with bone marrow-derived DCs and OT-I T cells induces DC activation, OT-I T cell proliferation, and antigen-specific T cell-mediated MC38 killing (A) HMGB1 secretion as measured in the supernatant collected from MC38 cancer cells treated with increasing concentrations of cisplatin (red) or carboplatin (black). Supernatant was collected 3 days after treatment. Data depict one representative experiment of two independent experiments; duplicate conditions for each experiment. Data are mean ± SEM. (B) Protein expression of MHC-I, PD-L1, CD40, and CD86 as measured by median fluorescence intensity in monocyte-derived DCs after 24 h of co-culture with MC38 cancer cells that were primed with dose titrations of cisplatin (red) or carboplatin (black) for 24 h. Data depict one representative experiment of three independent experiments; duplicate conditions for each experiment. Data are mean ± SEM. (C) Flow cytometry histograms depicting OT-I T cell proliferation measured by CellTrace Blue dilution assay. MC38-OVA cells were pretreated with dose titrations of cisplatin or carboplatin for 24 h. OT-I T cells were co-cultured with pretreated MC38-OVA cells for 3 days in the presence or absence of monocyte-derived DCs before the proliferation assay. Data depict one representative experiment of two independent experiments. (D and E) Representative flow cytogram showing OT-I T cell-mediated MC38-OVA cancer cell killing (D) and the summarized percentage of OT-I T cell-mediated tumor cell death (E) as measured by 7-AAD and annexin V staining. MC38-OVA tumor cells were pretreated with increasing concentrations of cisplatin or carboplatin for 24 h, followed by co-culture with OT-I T cells for 5 h before the assay. Data depict one representative experiment of three independent experiments; duplicate conditions for each experiment. Data are mean ± SEM. See also Figure S9.

References

    1. Galsky M.D., Chen G.J., Oh W.K., Bellmunt J., Roth B.J., Petrioli R., Dogliotti L., Dreicer R., Sonpavde G. Comparative effectiveness of cisplatin-based and carboplatin-based chemotherapy for treatment of advanced urothelial carcinoma. Ann. Oncol. 2012;23:406–410. - PubMed
    1. De Santis M., Bellmunt J., Mead G., Kerst J.M., Leahy M., Maroto P., Gil T., Marreaud S., Daugaard G., Skoneczna I., et al. Randomized phase II/III trial assessing gemcitabine/carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer who are unfit for cisplatin-based chemotherapy: EORTC study 30986. J. Clin. Oncol. 2012;30:191–199. - PMC - PubMed
    1. Sternberg C.N., de Mulder P., Schornagel J.H., Theodore C., Fossa S.D., van Oosterom A.T., Witjes J.A., Spina M., van Groeningen C.J., Duclos B., et al. Seven year update of an EORTC phase III trial of high-dose intensity M-VAC chemotherapy and G-CSF versus classic M-VAC in advanced urothelial tract tumours. Eur. J. Cancer. 2006;42:50–54. - PubMed
    1. Dash A., Galsky M.D., Vickers A.J., Serio A.M., Koppie T.M., Dalbagni G., Bochner B.H. Impact of renal impairment on eligibility for adjuvant cisplatin-based chemotherapy in patients with urothelial carcinoma of the bladder. Cancer. 2006;107:506–513. - PubMed
    1. Galsky M.D., Hahn N.M., Rosenberg J., Sonpavde G., Hutson T., Oh W.K., Dreicer R., Vogelzang N., Sternberg C.N., Bajorin D.F., Bellmunt J. Treatment of patients with metastatic urothelial cancer "unfit" for Cisplatin-based chemotherapy. J. Clin. Oncol. 2011;29:2432–2438. - PubMed

Publication types

MeSH terms

Associated data