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 Mar;30(3):716-729.
doi: 10.1038/s41591-024-02808-y. Epub 2024 Feb 13.

Biomarker-directed targeted therapy plus durvalumab in advanced non-small-cell lung cancer: a phase 2 umbrella trial

Affiliations
Clinical Trial

Biomarker-directed targeted therapy plus durvalumab in advanced non-small-cell lung cancer: a phase 2 umbrella trial

Benjamin Besse et al. Nat Med. 2024 Mar.

Abstract

For patients with non-small-cell lung cancer (NSCLC) tumors without currently targetable molecular alterations, standard-of-care treatment is immunotherapy with anti-PD-(L)1 checkpoint inhibitors, alone or with platinum-doublet therapy. However, not all patients derive durable benefit and resistance to immune checkpoint blockade is common. Understanding mechanisms of resistance-which can include defects in DNA damage response and repair pathways, alterations or functional mutations in STK11/LKB1, alterations in antigen-presentation pathways, and immunosuppressive cellular subsets within the tumor microenvironment-and developing effective therapies to overcome them, remains an unmet need. Here the phase 2 umbrella HUDSON study evaluated rational combination regimens for advanced NSCLC following failure of anti-PD-(L)1-containing immunotherapy and platinum-doublet therapy. A total of 268 patients received durvalumab (anti-PD-L1 monoclonal antibody)-ceralasertib (ATR kinase inhibitor), durvalumab-olaparib (PARP inhibitor), durvalumab-danvatirsen (STAT3 antisense oligonucleotide) or durvalumab-oleclumab (anti-CD73 monoclonal antibody). Greatest clinical benefit was observed with durvalumab-ceralasertib; objective response rate (primary outcome) was 13.9% (11/79) versus 2.6% (5/189) with other regimens, pooled, median progression-free survival (secondary outcome) was 5.8 (80% confidence interval 4.6-7.4) versus 2.7 (1.8-2.8) months, and median overall survival (secondary outcome) was 17.4 (14.1-20.3) versus 9.4 (7.5-10.6) months. Benefit with durvalumab-ceralasertib was consistent across known immunotherapy-refractory subgroups. In ATM-altered patients hypothesized to harbor vulnerability to ATR inhibition, objective response rate was 26.1% (6/23) and median progression-free survival/median overall survival were 8.4/22.8 months. Durvalumab-ceralasertib safety/tolerability profile was manageable. Biomarker analyses suggested that anti-PD-L1/ATR inhibition induced immune changes that reinvigorated antitumor immunity. Durvalumab-ceralasertib is under further investigation in immunotherapy-refractory NSCLC.ClinicalTrials.gov identifier: NCT03334617.

PubMed Disclaimer

Conflict of interest statement

B.B. has received grants or funds from Abbvie, Amgen, AstraZeneca, Chugai Pharmaceutical, Daiichi Sankyo, Ellipse Pharma, EISAI, Genmab, Genzyme Corporation, Hedera Dx, Inivata, IPSEN, Janssen, MSD, Pharmamar, Roche-Genentech, Sanofi, Socar Research, Tahio Oncology and Turning Point Therapeutics. E.P.-T. has participated in advisory councils or committees for AstraZeneca, Bristol Myers Squibb, Takeda and Sanofi. K.P. has participated in an advisory council or committee (steering committee) for and received grants or funds (out of this study) from AstraZeneca. S.H. has participated in advisory boards for MSD, Roche and AstraZeneca, and has received institutional grants or funds from AstraZeneca, GSK, Chiesi and Menarini Pharma. P.M.F. has received consulting fees from AstraZeneca, Bristol Myers Squibb, Novartis, Merck, Genentech, G1, F Star, Regeneron, Janssen, Sanofi, Amgen, Fosun, Teva, Synthekine, Flame, Iteos and Tavotek, and grants or funds from AstraZeneca, Bristol Myers Squibb, Regeneron, Novartis and BioNTech. M.J.H. has participated in advisory councils or committees for Bristol Myers Squibb, AstraZeneca, MSD, Lilly and Roche. M.M.A. has performed consulting or advisory roles with Merck, Pfizer, Bristol Myers Squibb, Foundation Medicine, Novartis, Gritstone Oncology, Mirati Therapeutics, EMD Serono, AstraZeneca, Instil Bio, Regeneron and Janssen, and has received institutional research funding from Genentech/Roche, Lilly, AstraZeneca, Bristol Myers Squibb, Amgen and Affini-T Therapeutics. M.T. has participated in advisory councils or committees for and received honoraria and consulting fees from AstraZeneca, Roche, Bristol Myers Squibb, MSD, Novartis, Sanofi, Takeda, Pfizer, Merck, Daiichi Sankyo, Janssen, Lilly and Boehringer Ingelheim, and has received grants or funds from AstraZeneca, Roche and Takeda. G.G. has received honoraria from AstraZeneca. P.W.-P. has participated in advisory councils or committees for AstraZeneca, Bristol Myers Squibb, Sanofi, Roche, Novartis, Jazz Pharmaceuticals and Pfizer, and has received honoraria from Bayer, Sanofi and Merck. F.A.S. has participated in advisory boards for and received honoraria and consulting fees from AstraZeneca, and their institution has received clinical trial payments from AstraZeneca. M.F. has participated in advisory councils or committees for AstraZeneca, Bristol Myers Squibb and Takeda, has received honoraria from AstraZeneca and Bristol Myers Squibb, and has received grants or funds from AstraZeneca. P.C. has participated in advisory councils or committees for AstraZeneca and Novartis, received honoraria from AstraZeneca, Janssen, Eli Lilly, GSK, Merck, Sanofi and Amgen, and received consulting fees from AstraZeneca, Bristol Myers Squibb, Roche, Amgen, Pfizer, Novartis and Merck. Q.S.C.C. has received honoraria and consulting fees from Abbvie, Amgen, AnHeart, AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Esperas Pharma, Eisai, Jazz Pharmaceutical, Janssen/Johnson and Johnson, Merck, Novartis, Ocellaris, Pfizer, Roche and Takeda. S.-W.K. has no conflicts of interest to declare. D.M. has received consulting fees from Abbvie, Arcus, Mirati and Lilly, and grants or funds from Merck, AstraZeneca, Incyte, Bristol Myers Squibb, Surface, Epicentrx, Boehringer Ingelheim, Y-mabs, Roche, Lilly, Pfizer and Novartis. M.L.J. has received institutional research funding from AbbVie, Acerta, Adaptimmune, Amgen, Apexigen, Arcus Biosciences, Array BioPharma, Artios Pharma, AstraZeneca, Atreca, BeiGene, BerGenBio, BioAtla, Black Diamond, Boehringer Ingelheim, Bristol Myers Squibb, Calithera Biosciences, Carisma Therapeutics, Checkpoint Therapeutics, City of Hope National Medical Center, Corvus Pharmaceuticals, Curis, CytomX, Daiichi Sankyo, Dracen Pharmaceuticals, Dynavax, Lilly, Elicio Therapeutics, EMD Serono, EQRx, Erasca, Exelixis, Fate Therapeutics, Genentech/Roche, Genmab, Genocea Biosciences, GlaxoSmithKline, Gritstone Oncology, Guardant Health, Harpoon, Helsinn Healthcare SA, Hengrui Therapeutics, Hutchison MediPharma, IDEAYA Biosciences, IGM Biosciences, Immunitas Therapeutics, Immunocore, Incyte, Janssen, Jounce Therapeutics, Kadmon Pharmaceuticals, Kartos Therapeutics, Loxo Oncology, Lycera, Memorial Sloan-Kettering, Merck, Merus, Mirati Therapeutics, Mythic Therapeutics, NeoImmune Tech, Neovia Oncology, Novartis, Numab Therapeutics, Nuvalent, OncoMed Pharmaceuticals, Palleon Pharmaceuticals, Pfizer, PMV Pharmaceuticals, Rain Therapeutics, RasCal Therapeutics, Regeneron Pharmaceuticals, Relay Therapeutics, Revolution Medicines, Ribon Therapeutics, Rubius Therapeutics, Sanofi, Seven and Eight Biopharmaceuticals/Birdie Biopharmaceuticals, Shattuck Labs, Silicon Therapeutics, Stem CentRx, Syndax Pharmaceuticals, Takeda Pharmaceuticals, Tarveda, TCR2 Therapeutics, Tempest Therapeutics, Tizona Therapeutics, TMUNITY Therapeutics, Turning Point Therapeutics, University of Michigan, Vyriad, WindMIL Therapeutics and Y-mAbs Therapeutics; and has received institutional payments for consulting or advisory roles for AbbVie, Amgen, Arcus Biosciences, Arrivent, Astellas, AstraZeneca, Black Diamond, Boehringer Ingelheim, Calithera Biosciences, Daiichi Sankyo, EcoR1, Genentech/Roche, Genmab, Genocea Biosciences, Gilead Sciences, GlaxoSmithKline, Gritstone Oncology, Ideaya Biosciences, Immunocore, iTeos, Janssen, Jazz Pharmaceuticals, Merck, Mirati Therapeutics, Molecular Axiom, Normunity, Novartis, Oncorus, Pyramid Biosciences, Regeneron Pharmaceuticals, Revolution Medicines, Sanofi-Aventis, SeaGen, Synthekine, Takeda Pharmaceuticals, Turning Point Therapeutics and VBL Therapeutics. S.C. has participated in advisory councils or committees for AstraZeneca, Bristol Myers Squibb, MSD and Sanofi. D.-W.K. has received research funding to their institution from Alpha Biopharma, Amgen, AstraZeneca/Medimmune, Boehringer Ingelheim, Bridge BioTherapeutics, Chong Keun Dang, Daiichi Sankyo, GSK, Hanmi, InnoN, Janssen, Merck, Merus, Mirati Therapeutics, MSD, Novartis, ONO Pharmaceutical, Pfizer, Roche/Genentech, Takeda, TP Therapeutics, Xcovery and Yuhan, and has received medical writing assistance from Amgen, AstraZeneca, Boehringer Ingelheim, Bridge BioTherapeutics, Chong Keun Dang, Daiichi Sankyo, GSK, Janssen, Merus, Mirati Therapeutics, MSD, Meck, Novartis, Pfizer, Roche, Takeda and Yuhan. M.T.M. has participated in advisory councils or committees for MSD, Takeda, Bayer, Amgen and Novartis, has received honoraria from Bristol Myers Squibb, MSD, Takeda, AstraZeneca, Roche and AbbVie, has received consulting fees from MSD and AstraZeneca, and has received grants or funds from AstraZeneca. D.V. has received honoraria from Bristol Myers Squibb, AstraZeneca, Pfizer, Novartis, Roche and Daichii. B.A., R.H., H.J.A., S.K., A.R., D.L.R., M.R.K., J.P.C., J.C.B., E.D., R.K., M.D., P.J.J., S.I., S.T.B. and J.C. are employees of and may own stock/shares in AstraZeneca. D.L.R. owns stocks or shares in Myriad Genetics. J.V.H. has participated in advisory councils or committees for AstraZeneca, Bristol Myers Squibb, Genentech/Roche, EMD Serono, GlaxoSmithKline, Hengrui Therapeutics, Eli Lilly, Novartis, Spectrum, Sanofi, Takeda, Mirati Therapeutics, Janssen Global Services and Leads Biolabs, has received honoraria from AstraZeneca, Bristol Myers Squibb, Genentech/Roche, EMD Serono, GlaxoSmithKline, Hengrui Therapeutics, Eli Lilly, Novartis, Spectrum, Sanofi, Takeda, Mirati Therapeutics, Janssen Global Services, Leads Biolabs and Boehringer Ingelheim, has received grants or funds from AstraZeneca, Spectrum, Boehringer Ingelheim and Takeda, and has received licensing fees from Spectrum.

Figures

Fig. 1
Fig. 1. CONSORT diagram of patient screening and disposition in HUDSON.
Based on tumor molecular profiling, patients were assigned to either biomarker-matched (Group A) or non-matched cohorts (Group B), which included patients with primary and acquired resistance determined by their initial response to prior immunotherapy-containing regimens. Patients were then treated with one of four durvalumab-based combination regimens (Modules 1–4).
Fig. 2
Fig. 2. PFS and OS with durvalumab–ceralasertib and with durvalumab plus olaparib, danvatirsen or oleclumab in HUDSON.
a,b, Kaplan–Meier analysis of PFS (a) and OS (b) among all patients who received durvalumab–ceralasertib or who received durvalumab plus olaparib, danvatirsen or oleclumab on HUDSON.
Fig. 3
Fig. 3. OS and gene expression profiling in subgroups known to be associated with poor immunotherapy response among patients who received durvalumab plus olaparib, danvatirsen or oleclumab.
af, Kaplan–Meier distributions of OS (left) and TIS ssGSEA enrichment score (ES) distributions (violin plots, right) comparing patients with primary or acquired resistance (a), PD-L1-negative or PD-L1-positive status (b), squamous cell carcinoma or adenocarcinoma histology (c), with or without bone/liver metastases (d), high or low TMB (e) and STK11-altered or STK11-wild-type tumors (f). Median OS, 80% CIs and numbers of patients at risk are shown in each Kaplan–Meier plot. Violin plots show data from individual patients (dots), color-coded by site of sample, the median value (text box) and the probability density distribution smoothed using a kernel density estimator (shaded areas). ssGSEA ES distributions were compared using two-sided Wilcoxon rank-sum tests, with no correction for multiplicity of testing; associated P values and the number of tumor samples in each group are reported for each comparison.
Fig. 4
Fig. 4. PFS and OS with durvalumab–ceralasertib and with durvalumab plus olaparib, danvatirsen or oleclumab in HUDSON in patient subgroups.
Figure shows medians and 80% CI per Kaplan–Meier methodology.
Fig. 5
Fig. 5. Ceralasertib induces systemic immunomodulation that enhances the immune-mediated effects of durvalumab in patients with NSCLC with progression on prior anti-PD-(L)1 treatment.
a, Schema depicting treatment schedule for modules 1 (durvalumab–olaparib), 2 (durvalumab–danvatirsen) and 3 (durvalumab–ceralasertib), showing durvalumab administration (blue arrows), combination agent dosing (continuous colored bars), and pretreatment/on-treatment blood sample collection time points (stars). BID, twice daily. bg, Effects of ceralasertib and durvalumab treatment on longitudinal blood-derived transcriptomes are depicted in the box plots showing differentially expressed monocyte-associated signatures (b), CD8 T-cell-associated signatures (c), dysfunctional/exhausted CD8 T-cell-associated signatures (d), TNF-α-associated signatures (e), interferon-γ-associated signatures (f) and interferon-α-associated signatures (g). P values are for comparisons with respective cycle 0, day (D) 1 time points. P values for visit dates represent linear mixed model effects, illustrating changes in on-treatment samples. hq, Effects of ceralasertib, olaparib and danvatirsen with durvalumab on longitudinal blood-derived T cell repertoire: box plots showing productive Simpson T cell clonality with durvalumab–ceralasertib (h); waterfall plots showing changes in clonality on treatment by patient with durvalumab–ceralasertib on cycle 0, day 1 versus cycle 1, day 1 (i) and cycle 1, day 1 versus cycle 1, day 22 (j); box plots showing distributions of T cell clonality with durvalumab–olaparib (k), and waterfall plots showing changes in clonality on treatment by patient with durvalumab–olaparib on cycle 1, day 1 versus cycle 1, day 15 (l); box plots showing distributions of T cell clonality with durvalumab–danvatirsen (m), and waterfall plots showing changes in clonality on treatment per patient with durvalumab–danvatirsen on cycle 1, day 1 versus cycle 1, day 15 (n); box plots of expanded T cell clones (o), newly detected expanded T cell clones (p) and Morisita index for durvalumab plus olaparib, danvatirsen or ceralasertib at indicated visit dates (q). P values for visit dates represent two-sided Wilcoxon paired signed-rank tests between time points or two-sided Wilcoxon signed-rank tests between study modules, with correction for multiplicity of testing (Benjamini–Hochberg procedure), illustrating changes in on-treatment samples. r, Proposed immunomodulatory mechanism of action effects of ceralasertib with durvalumab in the periphery and tumor. For all box plots (bh, k, m and oq), box centerlines show medians, box limits show upper and lower quartiles, and whiskers show range.
Fig. 5
Fig. 5. Ceralasertib induces systemic immunomodulation that enhances the immune-mediated effects of durvalumab in patients with NSCLC with progression on prior anti-PD-(L)1 treatment.
a, Schema depicting treatment schedule for modules 1 (durvalumab–olaparib), 2 (durvalumab–danvatirsen) and 3 (durvalumab–ceralasertib), showing durvalumab administration (blue arrows), combination agent dosing (continuous colored bars), and pretreatment/on-treatment blood sample collection time points (stars). BID, twice daily. bg, Effects of ceralasertib and durvalumab treatment on longitudinal blood-derived transcriptomes are depicted in the box plots showing differentially expressed monocyte-associated signatures (b), CD8 T-cell-associated signatures (c), dysfunctional/exhausted CD8 T-cell-associated signatures (d), TNF-α-associated signatures (e), interferon-γ-associated signatures (f) and interferon-α-associated signatures (g). P values are for comparisons with respective cycle 0, day (D) 1 time points. P values for visit dates represent linear mixed model effects, illustrating changes in on-treatment samples. hq, Effects of ceralasertib, olaparib and danvatirsen with durvalumab on longitudinal blood-derived T cell repertoire: box plots showing productive Simpson T cell clonality with durvalumab–ceralasertib (h); waterfall plots showing changes in clonality on treatment by patient with durvalumab–ceralasertib on cycle 0, day 1 versus cycle 1, day 1 (i) and cycle 1, day 1 versus cycle 1, day 22 (j); box plots showing distributions of T cell clonality with durvalumab–olaparib (k), and waterfall plots showing changes in clonality on treatment by patient with durvalumab–olaparib on cycle 1, day 1 versus cycle 1, day 15 (l); box plots showing distributions of T cell clonality with durvalumab–danvatirsen (m), and waterfall plots showing changes in clonality on treatment per patient with durvalumab–danvatirsen on cycle 1, day 1 versus cycle 1, day 15 (n); box plots of expanded T cell clones (o), newly detected expanded T cell clones (p) and Morisita index for durvalumab plus olaparib, danvatirsen or ceralasertib at indicated visit dates (q). P values for visit dates represent two-sided Wilcoxon paired signed-rank tests between time points or two-sided Wilcoxon signed-rank tests between study modules, with correction for multiplicity of testing (Benjamini–Hochberg procedure), illustrating changes in on-treatment samples. r, Proposed immunomodulatory mechanism of action effects of ceralasertib with durvalumab in the periphery and tumor. For all box plots (bh, k, m and oq), box centerlines show medians, box limits show upper and lower quartiles, and whiskers show range.
Extended Data Fig. 1
Extended Data Fig. 1. PFS and OS by treatment cohort.
PFS and OS with a, b, durvalumab-ceralasertib (median OS not yet mature in ATM biomarker-matched cohort), c, d, durvalumab-olaparib, e, f, durvalumab-danvatirsen, g, h, durvalumab-oleclumab.
Extended Data Fig. 2
Extended Data Fig. 2. Gene expression profiling in subgroups with known association with poor immunotherapy response among patients who received durvalumab plus olaparib, danvatirsen or oleclumab.
Gene-wise transcript-per-million (TPM) z-score heatmaps for the 18 genes comprising the tumour inflammation signature (TIS) in tumour samples from patients with a, primary or acquired resistance to prior immune checkpoint blockade therapy (n = 18, n = 31; overall prevalence: n = 81 [43.1%], n = 107 [56.9%]), b, PD-L1-positive or PD-L1-negative tumours (n = 16, n = 18; overall prevalence: n = 73 [58.9%], n = 51 [41.1%]), c, adenocarcinoma or squamous cell histology (n = 38, n = 6; overall prevalence: n = 130 [75.1%], n = 43 [24.9%]), d, with or without bone/liver metastases (n = 23, n = 26; overall prevalence: n = 76 [40.4%], n = 112 [59.6%]), e, high or low TMB (n = 16, n = 18; overall prevalence: n = 60 [41.1%], n = 86 [58.9%]) and f, STK11-altered or STK11-wild-type tumours (n = 5, n = 38; overall prevalence: n = 26 [14.9%], n = 148 [85.1%]). Tumour location and histology are annotated.
Extended Data Fig. 3
Extended Data Fig. 3. PFS and OS with durvalumab-ceralasertib and with durvalumab plus olaparib, danvatirsen or oleclumab in patient subgroups defined by presence or absence of adverse prognostic factors.
Kaplan–Meier analyses of (left) PFS and (right) OS in patients with a, primary resistance, b, acquired resistance, c, PD-L1-negative tumours, d, PD-L1-positive tumours, e, squamous histology, f, non-squamous histology, and g, STK11 mutations.
Extended Data Fig. 4
Extended Data Fig. 4. PFS and OS with durvalumab-ceralasertib in patient subgroups defined by presence or absence of adverse prognostic factors.
Kaplan–Meier analyses of (left) PFS and (right) OS with durvalumab-ceralasertib in patients with a, primary vs acquired resistance, b, PD-L1-negative vs -positive status, and c, squamous cell carcinoma vs non-squamous histology, and in patients d, with or without bone/liver metastases, e, with high or low tumour mutational burden, and according to f, STK11 status and g, KRAS status (variant or wild-type).

Comment in

References

    1. NCCN clinical practice guidelines in oncology: non-small cell lung cancer, version 3.2022. National Comprehensive Cancer Networkhttps://www.nccn.org/guidelines/guidelines-detail?category=1&id=1450 (2022). - PubMed
    1. Lamberti G, et al. Beyond EGFR, ALK and ROS1: current evidence and future perspectives on newly targetable oncogenic drivers in lung adenocarcinoma. Crit. Rev. Oncol. Hematol. 2020;156:103119. doi: 10.1016/j.critrevonc.2020.103119. - DOI - PubMed
    1. Sholl LM, et al. Multi-institutional oncogenic driver mutation analysis in lung adenocarcinoma: the Lung Cancer Mutation Consortium experience. J. Thorac. Oncol. 2015;10:768–777. doi: 10.1097/JTO.0000000000000516. - DOI - PMC - PubMed
    1. Lam VK, et al. Targeted tissue and cell-free tumor DNA sequencing of advanced lung squamous-cell carcinoma reveals clinically significant prevalence of actionable alterations. Clin. Lung Cancer. 2019;20:30–36 e33. doi: 10.1016/j.cllc.2018.08.020. - DOI - PubMed
    1. Brahmer J, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med. 2015;373:123–135. doi: 10.1056/NEJMoa1504627. - DOI - PMC - PubMed

Publication types

Associated data