Skip to main content
Top
Published in: Discover Oncology 1/2023

Open Access 01-12-2023 | Targeted Therapy | Research

A target map of clinical combination therapies in oncology: an analysis of clinicaltrials.gov

Authors: Jing Yang, Heming Kang, Liyang Lyu, Wei Xiong, Yuanjia Hu

Published in: Discover Oncology | Issue 1/2023

Login to get access

Abstract

Combination therapies have taken center stage for cancer treatment, however, there is a lack of a comprehensive portrait to quantitatively map the current clinical combination progress. This study aims to capture clinical combination therapies of the validated FDA-approved new oncology drugs by a macro data analysis and to summarize combination mechanisms and strategies in the context of the existing literature. A total of 72 new molecular entities or new therapeutic biological products for cancer treatment approved by the FDA from 2017 to 2021 were identified, and the data on their related 3334 trials were retrieved from the database of ClinicalTrials.gov. Moreover, these sampled clinical trials were refined by activity status and combination relevance and labeled with the relevant clinical arms and drug combinations, as well as drug targets and target pairs. Combination therapies are increasingly prevalent in clinical trials of new oncology drugs. From retrospective work, existing clinical combination therapies in oncology are driven by different patterns (i.e., rational design and industry trends). The former can be represented by mechanism-based or structure-based combinations, such as targeting different domains of HER2 protein or in-series co-targeting in RAF plus MEK inhibitors. The latter is an empirically driven strategy, including redundant combinations in hot targets, such as PD-1/PD-L1, PI3K, CDK4/6, and PARP. Because of an explosion in the number of clinical trials and the resultant shortage of available patients, it is essential to rationally design drug combinations.
Appendix
Available only for authorised users
Literature
2.
go back to reference Bonavida B, Chouaib S. Resistance to anticancer immunity in cancer patients: potential strategies to reverse resistance. Ann Oncol. 2017;28:457–67.PubMedCrossRef Bonavida B, Chouaib S. Resistance to anticancer immunity in cancer patients: potential strategies to reverse resistance. Ann Oncol. 2017;28:457–67.PubMedCrossRef
3.
go back to reference Fitzgerald JB, Schoeberl B, Nielsen UB, et al. Systems biology and combination therapy in the quest for clinical efficacy. Nat Chem Biol. 2006;2:458–66.PubMedCrossRef Fitzgerald JB, Schoeberl B, Nielsen UB, et al. Systems biology and combination therapy in the quest for clinical efficacy. Nat Chem Biol. 2006;2:458–66.PubMedCrossRef
4.
go back to reference Boshuizen J, Peeper DS. Rational cancer treatment combinations: an urgent clinical need. Mol Cell. 2020;78:1002–18.PubMedCrossRef Boshuizen J, Peeper DS. Rational cancer treatment combinations: an urgent clinical need. Mol Cell. 2020;78:1002–18.PubMedCrossRef
5.
go back to reference de Miguel M, Calvo E. Clinical challenges of immune checkpoint inhibitors. Cancer Cell. 2020;38:326–33.PubMedCrossRef de Miguel M, Calvo E. Clinical challenges of immune checkpoint inhibitors. Cancer Cell. 2020;38:326–33.PubMedCrossRef
6.
go back to reference Tonekaboni SAM, Ghoraie LS, Manem VSK, et al. Predictive approaches for drug combination discovery in cancer. Brief Bioinform. 2018;19:263–76.CrossRef Tonekaboni SAM, Ghoraie LS, Manem VSK, et al. Predictive approaches for drug combination discovery in cancer. Brief Bioinform. 2018;19:263–76.CrossRef
7.
go back to reference Schmidt EV, Chisamore MJ, Chaney MF, et al. Assessment of clinical activity of pd-1 checkpoint inhibitor combination therapies reported in clinical trials. Jama Netw Open. 2020;3:e1920833.PubMedCrossRef Schmidt EV, Chisamore MJ, Chaney MF, et al. Assessment of clinical activity of pd-1 checkpoint inhibitor combination therapies reported in clinical trials. Jama Netw Open. 2020;3:e1920833.PubMedCrossRef
8.
go back to reference Palmer AC, Sorger PK. Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell. 2017;171:1678-91.e1613.PubMedPubMedCentralCrossRef Palmer AC, Sorger PK. Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell. 2017;171:1678-91.e1613.PubMedPubMedCentralCrossRef
10.
go back to reference Wu M, Huang Q, Xie Y, et al. Improvement of the anticancer efficacy of pd-1/pd-l1 blockade via combination therapy and pd-l1 regulation. J Hematol Oncol. 2022;15:24.PubMedPubMedCentralCrossRef Wu M, Huang Q, Xie Y, et al. Improvement of the anticancer efficacy of pd-1/pd-l1 blockade via combination therapy and pd-l1 regulation. J Hematol Oncol. 2022;15:24.PubMedPubMedCentralCrossRef
11.
go back to reference Vinciguerra GLR, Sonego M, Segatto I, et al. Cdk4/6 inhibitors in combination therapies: better in company than alone: a mini review. Front Oncol. 2022;12:891580.CrossRef Vinciguerra GLR, Sonego M, Segatto I, et al. Cdk4/6 inhibitors in combination therapies: better in company than alone: a mini review. Front Oncol. 2022;12:891580.CrossRef
12.
go back to reference Drean A, Lord CJ, Ashworth A. Parp inhibitor combination therapy. Crit Rev Oncol Hematol. 2016;108:73–85.PubMedCrossRef Drean A, Lord CJ, Ashworth A. Parp inhibitor combination therapy. Crit Rev Oncol Hematol. 2016;108:73–85.PubMedCrossRef
13.
14.
go back to reference Salama AKS, Moschos SJ. Next steps in immuno-oncology: enhancing antitumor effects through appropriate patient selection and rationally designed combination strategies. Ann Oncol. 2017;28:57–74.PubMedCrossRef Salama AKS, Moschos SJ. Next steps in immuno-oncology: enhancing antitumor effects through appropriate patient selection and rationally designed combination strategies. Ann Oncol. 2017;28:57–74.PubMedCrossRef
15.
go back to reference Iafolla MAJ, Selby H, Warner K, et al. Rational design and identification of immuno-oncology drug combinations. Eur J Cancer. 2018;95:38–51.PubMedCrossRef Iafolla MAJ, Selby H, Warner K, et al. Rational design and identification of immuno-oncology drug combinations. Eur J Cancer. 2018;95:38–51.PubMedCrossRef
16.
go back to reference Dudani S, Graham J, Wells JC, et al. First-line immuno-oncology combination therapies in metastatic renal-cell carcinoma: results from the international metastatic renal-cell carcinoma database consortium. Eur Urol. 2019;76:861–7.PubMedPubMedCentralCrossRef Dudani S, Graham J, Wells JC, et al. First-line immuno-oncology combination therapies in metastatic renal-cell carcinoma: results from the international metastatic renal-cell carcinoma database consortium. Eur Urol. 2019;76:861–7.PubMedPubMedCentralCrossRef
17.
go back to reference Maione P, Gridelli C, Troiani T, et al. Combining targeted therapies and drugs with multiple targets in the treatment of nsclc. Oncologist. 2006;11:274–84.PubMedCrossRef Maione P, Gridelli C, Troiani T, et al. Combining targeted therapies and drugs with multiple targets in the treatment of nsclc. Oncologist. 2006;11:274–84.PubMedCrossRef
19.
go back to reference Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: treating cancer with specificity. Eur J Pharmacol. 2018;834:188–96.CrossRefPubMed Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: treating cancer with specificity. Eur J Pharmacol. 2018;834:188–96.CrossRefPubMed
20.
go back to reference Tsimberidou AM. Targeted therapy in cancer. Cancer Chemoth Pharm. 2015;76:1113–32.CrossRef Tsimberidou AM. Targeted therapy in cancer. Cancer Chemoth Pharm. 2015;76:1113–32.CrossRef
21.
go back to reference Kidera Y, Satoh T, Ueda S, et al. High-dose dexamethasone plus antihistamine prevents colorectal cancer patients treated with modified folfox6 from hypersensitivity reactions induced by oxaliplatin. Int J Clin Oncol. 2011;16:244–9.PubMedCrossRef Kidera Y, Satoh T, Ueda S, et al. High-dose dexamethasone plus antihistamine prevents colorectal cancer patients treated with modified folfox6 from hypersensitivity reactions induced by oxaliplatin. Int J Clin Oncol. 2011;16:244–9.PubMedCrossRef
22.
go back to reference Kang RY, Yoo KS, Han HJ, et al. Evaluation of the effects and adverse drug reactions of low-dose dexamethasone premedication with weekly docetaxel. Support Care Cancer. 2017;25:429–37.PubMedCrossRef Kang RY, Yoo KS, Han HJ, et al. Evaluation of the effects and adverse drug reactions of low-dose dexamethasone premedication with weekly docetaxel. Support Care Cancer. 2017;25:429–37.PubMedCrossRef
23.
go back to reference Aldea M, Orillard E, Mansi L, et al. How to manage patients with corticosteroids in oncology in the era of immunotherapy? Eur J Cancer. 2020;141:239–51.PubMedCrossRef Aldea M, Orillard E, Mansi L, et al. How to manage patients with corticosteroids in oncology in the era of immunotherapy? Eur J Cancer. 2020;141:239–51.PubMedCrossRef
24.
go back to reference Janowitz T, Kleeman S, Vonderheide RH. Reconsidering dexamethasone for antiemesis when combining chemotherapy and immunotherapy. Oncologist. 2021;26:269–73.PubMedPubMedCentralCrossRef Janowitz T, Kleeman S, Vonderheide RH. Reconsidering dexamethasone for antiemesis when combining chemotherapy and immunotherapy. Oncologist. 2021;26:269–73.PubMedPubMedCentralCrossRef
25.
go back to reference Giles AJ, Hutchinson MKND, Sonnemann HM, et al. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer. 2018;6:51.PubMedPubMedCentralCrossRef Giles AJ, Hutchinson MKND, Sonnemann HM, et al. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer. 2018;6:51.PubMedPubMedCentralCrossRef
26.
go back to reference Arbour KC, Mezquita L, Long N, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol. 2018;36:2872–8.PubMedCrossRef Arbour KC, Mezquita L, Long N, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol. 2018;36:2872–8.PubMedCrossRef
29.
go back to reference Wang DX, Lin JZ, Yang X, et al. Combination regimens with pd-1/pd-l1 immune checkpoint inhibitors for gastrointestinal malignancies. J Hematol Oncol. 2019;12:42.PubMedPubMedCentralCrossRef Wang DX, Lin JZ, Yang X, et al. Combination regimens with pd-1/pd-l1 immune checkpoint inhibitors for gastrointestinal malignancies. J Hematol Oncol. 2019;12:42.PubMedPubMedCentralCrossRef
30.
32.
go back to reference Huang MY, Jiang XM, Wang BL, et al. Combination therapy with pd-1/pd-l1 blockade in non-small cell lung cancer: strategies and mechanisms. Pharmacol Therapeut. 2021;219:107694.CrossRef Huang MY, Jiang XM, Wang BL, et al. Combination therapy with pd-1/pd-l1 blockade in non-small cell lung cancer: strategies and mechanisms. Pharmacol Therapeut. 2021;219:107694.CrossRef
33.
go back to reference Wang QH, Wu X. Primary and acquired resistance to pd-1/pd-l1 blockade in cancer treatment. Int Immunopharmacol. 2017;46:210–9.PubMedCrossRef Wang QH, Wu X. Primary and acquired resistance to pd-1/pd-l1 blockade in cancer treatment. Int Immunopharmacol. 2017;46:210–9.PubMedCrossRef
34.
go back to reference Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 2022;19:237–53.PubMedCrossRef Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 2022;19:237–53.PubMedCrossRef
37.
go back to reference Zhang YQ, Brekken RA. Direct and indirect regulation of the tumor immune microenvironment by vegf. J Leukocyte Biol. 2022;111:1269–86.PubMedCrossRef Zhang YQ, Brekken RA. Direct and indirect regulation of the tumor immune microenvironment by vegf. J Leukocyte Biol. 2022;111:1269–86.PubMedCrossRef
38.
go back to reference Kudo M. Scientific rationale for combined immunotherapy with pd-1/pd-l1 antibodies and vegf inhibitors in advanced hepatocellular carcinoma. Cancers. 2020;12:1089.PubMedPubMedCentralCrossRef Kudo M. Scientific rationale for combined immunotherapy with pd-1/pd-l1 antibodies and vegf inhibitors in advanced hepatocellular carcinoma. Cancers. 2020;12:1089.PubMedPubMedCentralCrossRef
39.
go back to reference Ramakrishnan S. Hif-2 in cancer-associated fibroblasts polarizes macrophages and creates an immunosuppressive tumor microenvironment in pancreatic cancer. Gastroenterology. 2022;162:1835–7.PubMedCrossRef Ramakrishnan S. Hif-2 in cancer-associated fibroblasts polarizes macrophages and creates an immunosuppressive tumor microenvironment in pancreatic cancer. Gastroenterology. 2022;162:1835–7.PubMedCrossRef
41.
go back to reference McGettrick AF, O’Neill LAJ. The role of hif in immunity and inflammation. Cell Metab. 2020;32:524–36.PubMedCrossRef McGettrick AF, O’Neill LAJ. The role of hif in immunity and inflammation. Cell Metab. 2020;32:524–36.PubMedCrossRef
42.
go back to reference Marshall LA, Marubayashi S, Jorapur A, et al. Tumors establish resistance to immunotherapy by regulating t(reg) recruitment via ccr4. J Immunother Cancer. 2020;8:e000764.PubMedPubMedCentralCrossRef Marshall LA, Marubayashi S, Jorapur A, et al. Tumors establish resistance to immunotherapy by regulating t(reg) recruitment via ccr4. J Immunother Cancer. 2020;8:e000764.PubMedPubMedCentralCrossRef
43.
44.
go back to reference Yonemitsu K, Pan C, Fujiwara Y, et al. Gm-csf derived from the inflammatory microenvironment potentially enhanced pd-l1 expression on tumor-associated macrophages in human breast cancer. Sci Rep. 2022;12:12007.PubMedPubMedCentralCrossRef Yonemitsu K, Pan C, Fujiwara Y, et al. Gm-csf derived from the inflammatory microenvironment potentially enhanced pd-l1 expression on tumor-associated macrophages in human breast cancer. Sci Rep. 2022;12:12007.PubMedPubMedCentralCrossRef
46.
go back to reference Oba T, Long MD, Keler T, et al. Overcoming primary and acquired resistance to anti-pd-l1 therapy by induction and activation of tumor-residing cdc1s. Nat Commun. 2020;11:5415.PubMedPubMedCentralCrossRef Oba T, Long MD, Keler T, et al. Overcoming primary and acquired resistance to anti-pd-l1 therapy by induction and activation of tumor-residing cdc1s. Nat Commun. 2020;11:5415.PubMedPubMedCentralCrossRef
49.
go back to reference Gu WX, Qu RB, Meng FH, et al. Polymeric nanomedicines targeting hematological malignancies. J Control Release. 2021;337:571–88.PubMedCrossRef Gu WX, Qu RB, Meng FH, et al. Polymeric nanomedicines targeting hematological malignancies. J Control Release. 2021;337:571–88.PubMedCrossRef
50.
go back to reference Xie BL, Li ZD, Zhou JF, et al. Current status and perspectives of dual-targeting chimeric antigen receptor t-cell therapy for the treatment of hematological malignancies. Cancers. 2022;14:3230.PubMedPubMedCentralCrossRef Xie BL, Li ZD, Zhou JF, et al. Current status and perspectives of dual-targeting chimeric antigen receptor t-cell therapy for the treatment of hematological malignancies. Cancers. 2022;14:3230.PubMedPubMedCentralCrossRef
51.
go back to reference Attwood MM, Fabbro D, Sokolov AV, et al. Trends in kinase drug discovery: targets, indications and inhibitor design. Nat Rev Drug Discov. 2021;20:839–61.PubMedCrossRef Attwood MM, Fabbro D, Sokolov AV, et al. Trends in kinase drug discovery: targets, indications and inhibitor design. Nat Rev Drug Discov. 2021;20:839–61.PubMedCrossRef
52.
53.
go back to reference Arpino G, Wiechmann L, Osborne CK, et al. Crosstalk between the estrogen receptor and the her tyrosine kinase receptor family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocr Rev. 2008;29:217–33.PubMedPubMedCentralCrossRef Arpino G, Wiechmann L, Osborne CK, et al. Crosstalk between the estrogen receptor and the her tyrosine kinase receptor family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocr Rev. 2008;29:217–33.PubMedPubMedCentralCrossRef
54.
go back to reference Lopez-Otin C, Hunter T. The regulatory crosstalk between kinases and proteases in cancer. Nat Rev Cancer. 2010;10:278–92.PubMedCrossRef Lopez-Otin C, Hunter T. The regulatory crosstalk between kinases and proteases in cancer. Nat Rev Cancer. 2010;10:278–92.PubMedCrossRef
55.
go back to reference Sun C, Bernards R. Feedback and redundancy in receptor tyrosine kinase signaling: relevance to cancer therapies. Trends Biochem Sci. 2014;39:465–74.PubMedCrossRef Sun C, Bernards R. Feedback and redundancy in receptor tyrosine kinase signaling: relevance to cancer therapies. Trends Biochem Sci. 2014;39:465–74.PubMedCrossRef
56.
go back to reference Scheiblecker L, Kollmann K, Sexl V. Cdk4/6 and mapk-crosstalk as opportunity for cancer treatment. Pharmaceuticals-Base. 2020;13:418.CrossRef Scheiblecker L, Kollmann K, Sexl V. Cdk4/6 and mapk-crosstalk as opportunity for cancer treatment. Pharmaceuticals-Base. 2020;13:418.CrossRef
57.
go back to reference Ding XK, Sharko AC, McDermott MSJ, et al. Inhibition of cdk8/19 mediator kinase potentiates her2-targeting drugs and bypasses resistance to these agents in vitro and in vivo. P Natl Acad Sci USA. 2022;119:e2201073119.CrossRef Ding XK, Sharko AC, McDermott MSJ, et al. Inhibition of cdk8/19 mediator kinase potentiates her2-targeting drugs and bypasses resistance to these agents in vitro and in vivo. P Natl Acad Sci USA. 2022;119:e2201073119.CrossRef
58.
go back to reference Recondo G, Bahcall M, Spurr LF, et al. Molecular mechanisms of acquired resistance to met tyrosine kinase inhibitors in patients with met exon 14-mutant nsclc. Clin Cancer Res. 2020;26:2615–25.PubMedCrossRef Recondo G, Bahcall M, Spurr LF, et al. Molecular mechanisms of acquired resistance to met tyrosine kinase inhibitors in patients with met exon 14-mutant nsclc. Clin Cancer Res. 2020;26:2615–25.PubMedCrossRef
59.
go back to reference Lai YC, Zhao ZJ, Zeng T, et al. Crosstalk between vegfr and other receptor tyrosine kinases for tki therapy of metastatic renal cell carcinoma. Cancer Cell Int. 2018;18:31.PubMedPubMedCentralCrossRef Lai YC, Zhao ZJ, Zeng T, et al. Crosstalk between vegfr and other receptor tyrosine kinases for tki therapy of metastatic renal cell carcinoma. Cancer Cell Int. 2018;18:31.PubMedPubMedCentralCrossRef
61.
go back to reference Karaman MW, Herrgard S, Treiber DK, et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–32.PubMedCrossRef Karaman MW, Herrgard S, Treiber DK, et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–32.PubMedCrossRef
64.
go back to reference Bradbury A, Hall S, Curtin N, et al. Targeting atr as cancer therapy: a new era for synthetic lethality and synergistic combinations? Pharmacol Therapeut. 2020;207:107450.CrossRef Bradbury A, Hall S, Curtin N, et al. Targeting atr as cancer therapy: a new era for synthetic lethality and synergistic combinations? Pharmacol Therapeut. 2020;207:107450.CrossRef
65.
go back to reference Hynes NE, Lane HA. Erbb receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5:341–54.PubMedCrossRef Hynes NE, Lane HA. Erbb receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5:341–54.PubMedCrossRef
66.
go back to reference Nahta R, Hung MC, Esteva FJ. The her-2-targeting antibodies trastuzumab and pertuzumab synergistically inhibit the survival of breast cancer cells. Cancer Res. 2004;64:2343–6.PubMedCrossRef Nahta R, Hung MC, Esteva FJ. The her-2-targeting antibodies trastuzumab and pertuzumab synergistically inhibit the survival of breast cancer cells. Cancer Res. 2004;64:2343–6.PubMedCrossRef
67.
go back to reference Shah M, Wedam S, Cheng J, et al. Fda approval summary: tucatinib for the treatment of patients with advanced or metastatic her2-positive breast cancer. Clin Cancer Res. 2021;27:1220–6.PubMedCrossRef Shah M, Wedam S, Cheng J, et al. Fda approval summary: tucatinib for the treatment of patients with advanced or metastatic her2-positive breast cancer. Clin Cancer Res. 2021;27:1220–6.PubMedCrossRef
71.
72.
go back to reference Yuan XR, Bu H, Zhou JP, et al. Recent advances of shp2 inhibitors in cancer therapy: current development and clinical application. J Med Chem. 2020;63:11368–96.PubMedCrossRef Yuan XR, Bu H, Zhou JP, et al. Recent advances of shp2 inhibitors in cancer therapy: current development and clinical application. J Med Chem. 2020;63:11368–96.PubMedCrossRef
75.
go back to reference Li ZW, Wang DD, Chen XT, et al. Prmt1-mediated ezh2 methylation promotes breast cancer cell proliferation and tumorigenesis. Cell Death Dis. 2021;12:1080.PubMedPubMedCentralCrossRef Li ZW, Wang DD, Chen XT, et al. Prmt1-mediated ezh2 methylation promotes breast cancer cell proliferation and tumorigenesis. Cell Death Dis. 2021;12:1080.PubMedPubMedCentralCrossRef
76.
go back to reference Yu JX, Hodge JP, Oliva C, et al. Trends in clinical development for pd-1/pd-l1 inhibitors. Nat Rev Drug Discov. 2020;19:163–4.CrossRef Yu JX, Hodge JP, Oliva C, et al. Trends in clinical development for pd-1/pd-l1 inhibitors. Nat Rev Drug Discov. 2020;19:163–4.CrossRef
77.
go back to reference Rotte A. Combination of ctla-4 and pd-1 blockers for treatment of cancer. J Exp Clin Canc Res. 2019;38:255.CrossRef Rotte A. Combination of ctla-4 and pd-1 blockers for treatment of cancer. J Exp Clin Canc Res. 2019;38:255.CrossRef
78.
go back to reference Grilley-Olson JE, Bedard PL, Fasolo A, et al. A phase ib dose-escalation study of the mek inhibitor trametinib in combination with the pi3k/mtor inhibitor gsk2126458 in patients with advanced solid tumors. Invest New Drug. 2016;34:740–9.CrossRef Grilley-Olson JE, Bedard PL, Fasolo A, et al. A phase ib dose-escalation study of the mek inhibitor trametinib in combination with the pi3k/mtor inhibitor gsk2126458 in patients with advanced solid tumors. Invest New Drug. 2016;34:740–9.CrossRef
80.
go back to reference Bauml J, Cho BC, Park K, et al. Amivantamab in combination with lazertinib for the treatment of osimertinib-relapsed, chemotherapy-naive egfr mutant (egfrm) non-small cell lung cancer (nsclc) and potential biomarkers for response. J Clin Oncol. 2021;39:9006.CrossRef Bauml J, Cho BC, Park K, et al. Amivantamab in combination with lazertinib for the treatment of osimertinib-relapsed, chemotherapy-naive egfr mutant (egfrm) non-small cell lung cancer (nsclc) and potential biomarkers for response. J Clin Oncol. 2021;39:9006.CrossRef
Metadata
Title
A target map of clinical combination therapies in oncology: an analysis of clinicaltrials.gov
Authors
Jing Yang
Heming Kang
Liyang Lyu
Wei Xiong
Yuanjia Hu
Publication date
01-12-2023
Publisher
Springer US
Published in
Discover Oncology / Issue 1/2023
Print ISSN: 1868-8497
Electronic ISSN: 2730-6011
DOI
https://doi.org/10.1007/s12672-023-00758-4

Other articles of this Issue 1/2023

Discover Oncology 1/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine