• No results found

University of Groningen Identification and modulation of drug targets for precision medicine in breast, lung and ovarian cancer subtypes Stutvoet, Thijs

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Identification and modulation of drug targets for precision medicine in breast, lung and ovarian cancer subtypes Stutvoet, Thijs"

Copied!
9
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Identification and modulation of drug targets for precision medicine in breast, lung and ovarian

cancer subtypes

Stutvoet, Thijs

DOI:

10.33612/diss.144705120

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Stutvoet, T. (2020). Identification and modulation of drug targets for precision medicine in breast, lung and

ovarian cancer subtypes. University of Groningen. https://doi.org/10.33612/diss.144705120

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Introduction

(3)

INTRODUCTION

The number of cancer patients is increasing worldwide due to aging of the general population and lifestyle-related risk factors, resulting in an estimated 18.1 million new cancer cases annually.1 Cancer has become the leading cause of death in the Western

world.2 Most cancers are treated with either surgery, radiotherapy, systemic therapy,

or a combination of these treatments. Systemic therapies, including chemotherapy, hormonal therapies, targeted agents, and immunotherapy, provide treatment options in the adjuvant and advanced-stage setting. Treatment failure is often caused by tumor resistance to therapy, resulting in disease progression and death.3

Chemotherapy preferentially kills cancer cells by targeting their increased proliferation and decreased recovery potential compared to healthy cells. Chemotherapy is applied as standard of care for patients with many advanced cancer types.4,5 Several biological

mechanisms that determine chemotherapy response have been discovered.3,4 More

recently, studies into the biology of cancer have resulted in the discovery of cancer drivers, which can now be directly targeted using antibodies and small molecule inhibitors.6–8 Targeted therapies that improve clinical outcomes for patients and have

become standard treatment, include the human epidermal growth factor receptor 2 (HER2) inhibitors trastuzumab and lapatinib in patients with HER2 overexpressing breast cancer, and the epidermal growth factor receptor (EGFR) inhibitor erlotinib in patients with EGFR mutant non-small cell lung cancer (NSCLC).9–11 However, most breast

cancers and NSCLCs do not harbor HER2 overexpression or EGFR mutations.7 Therefore,

additional tumor targets for therapy need to be uncovered to provide treatment options for a larger number of patients.

Large scale analyses, such as efforts by The Cancer Genome Atlas (TCGA) project assess mutations, and alterations in copy number, methylation and gene expression to define and characterize subgroups of cancers.12 Importantly, most of this data is accessible

through public repositories such as the Gene Expression Omnibus (GEO), enabling further research.13 These databases are steadily growing in volume and diversity,

allowing for identification of new cancer drivers and molecular cancer subtypes.14 For

example, this unveiled novel alterations in HER2, hepatocyte growth factor receptor, and guanosine triphosphate (GTP)-binding protein Ras Like Without CAAX 1 (RIT1) that are currently studied as treatment targets in NSCLC patients.15,16 Other cancer

subtype-related findings by the TCGA Research Network include the suggestion that over half the high grade serous ovarian cancers (HGSOC) harbor deficiencies in the homologous recombination pathway, greatly expanding the expected number of Poly (ADP-ribose)

1

10 Chapter 1

(4)

polymerase (PARP) inhibitor sensitive HGSOC tumors.17 Subsequent clinical studies

confirmed efficacy of PARP inhibitors as maintenance therapy following a response to platinum-based therapy in patients with HGSOC, which is now standard of care.18

Although targeted therapies can induce rapid and strong responses, efficacy is often limited by recurrences with therapy-resistant disease.7,19 This indicates a need for more

treatments with long-term efficacy.

Immunotherapy is a new treatment strategy that can induce long-term tumor responses in patients. These treatments include antibodies that block the function of immune checkpoint proteins, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death-1 (PD-1), and programmed death-ligand 1 (PD-L1), reinvigorating the anti-tumor immune response.20,21 In aggressive tumor types, including advanced

melanoma and NSCLC, immunotherapy can result in responses that persist after discontinuation of treatment.21,22 To improve immunotherapy response rates, patient

selection can be performed, but this is still in an early stage. For instance, tumor cell PD-L1 expression is used to select NSCLC patients for immune checkpoint inhibitor therapies. However, even from the patients with PD-L1 positive tumors 55% do not respond to these treatments, while 10% of patients with PD-L1 negative tumors respond.23–25 The

poor predictive value of tumor PD-L1 expression may be caused by transient PD-L1 expression or heterogeneity of PD-L1 expression within and between tumor lesions, none of which are taken into account when analyzing a single biopsy.26 Whole-body

imaging techniques, such as positron emission tomography (PET), may provide more insight in PD-L1 expression and improve patient selection for immunotherapy, as shown for PD-L1 targeted treatment in NSCLC, triple negative breast cancer (TNBC), and bladder cancer.27 Moreover, the non-invasive nature of PET imaging enables serial

analysis of target expression, allowing follow-up of PD-L1 expression in tumors. Currently, many trials are studying combination therapies in patients with various types of cancer to improve the efficacy of immunotherapy.28 Improved insight into immune

checkpoint biology may guide the development of new combination therapies including immunotherapy, targeted therapies, and chemotherapy.

AIM OF THIS THESIS

Providing a biological rationale for potential combination therapies and patient selection tools to enhance efficacy of immunotherapy and other targeted therapies in patients with subtypes of breast cancer, NSCLC, and ovarian cancer.

(5)

GENERAL OUTLINE OF THIS THESIS

Breast cancer subtypes harbor diverse underlying biology, enabling the use of subtype-specific therapies. In chapter 2, we review knowledge regarding immunotherapy in

breast cancer. We perform a literature search including PubMed and the ClinicalTrials. gov database and abstracts from the American Society of Clinical Oncology (ASCO) annual meetings, San Antonio Breast Cancer symposium, American Association of Cancer Research (AACR) annual meeting and the annual congresses of the European Society of Medical Oncology (ESMO). We focus on the differential immunogenicity of breast cancer subtypes and prognostic value of tumor infiltrating lymphocytes and immune checkpoints. Furthermore, we summarize the immunological effects of current therapies and describe upcoming additional immunotherapeutic treatment strategies in breast cancer.

Patients with NCSLCs without targetable driver mutations respond better to PD-1/ PD-L1 targeted immunotherapy. In chapter 3, we investigate regulation of PD-L1 in

this subtype of NSCLC in the context of EGFR and interferon gamma (IFNγ) signaling. Using data from the TCGA and validated pathway activation signatures, we search for associations between activation of downstream EGFR and IFNγ pathways and PD-L1 gene expression. Next, we validate our findings at the mRNA and protein level in lung adenocarcinoma cell lines using ligands and specific inhibitors. Additionally, we study the mechanistic basis of PD-L1 regulation in these cell lines using flow cytometry, Western blotting, MTT assays, and quantitative PCR.

For certain cancer types, PD-L1 immunohistochemistry is used to select patients for PD-1/PD-L1 targeted therapies, but performance of this biomarker is suboptimal. In

chapter 4, we evaluate performance of a human-PD-L1-specific PET tracer. This

adnectin-based, fluorine-18 labeled tracer, allows imaging on the day of administration. We characterize the tracer using human lung and ovarian cancer cell lines with different basal PD-L1 expression levels, and study binding and specificity of the tracer. Moreover, we evaluated in vivo usability of the tracer to detect basal PD-L1 expression levels and IFNγ- and the mitogen-activated protein kinase 1 and 2 (MEK1/2) inhibitor selumetinib-induced changes of PD-L1 expression levels using xenograft models of these cell lines. Despite the introduction of PARP and angiogenesis inhibitors, overall survival of patients with advanced high grade serous ovarian cancer remains poor. In chapter 5, we use

independent component analysis to extract the transcriptomic footprints of biological signals from publicly available gene expression data from 1,125 epithelial ovarian cancer

1

12 Chapter 1

(6)

samples, spanning all different histological subsets. We link known and unknown biology of the epithelial ovarian cancer subtypes to the uncovered transcriptomic footprints and relate these to prognosis of patients with platinum-treated serous ovarian cancer using survival tree analysis to uncover subtype-specific targets for therapy.

Patients with advanced ovarian clear cell cancer have worse survival compared to patients with other ovarian cancer subtypes due to the platinum resistant nature of their disease. In chapter 6, we look deeper into targeted therapies for ovarian clear cell

cancer. Using publicly available gene expression data from normal ovarian epithelium and clear cell ovarian cancer samples, we uncover enrichment for DNA repair signaling. Using MTT and clonogenic assays we study sensitivity of clear cell ovarian cancer cell lines to inhibition of the DNA damage response and cisplatin. To explain our findings, we look deeper into the induction and resolution of DNA damage and apoptosis after exposure to cisplatin using flow cytometry and immune fluorescence.

The findings of this thesis and the future perspectives are summarized in chapter 7. Chapter 8 provides a summary in Dutch.

(7)

REFERENCES

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394-424. doi:10.3322/caac.21492.

2. Dagenais GR, Leong DP, Rangarajan S, et al. Variations in common diseases, hospital admissions, and deaths in middle-aged adults in 21 countries from five continents (PURE): a prospective cohort study.

Lancet. 2019;6736(19):1-10. doi:10.1016/S0140-6736(19)32007-0.

3. Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 2007;7(8):573-584. doi:10.1038/nrc2167.

4. Galmarini D, Galmarini CM, Galmarini FC. Cancer chemotherapy: A critical analysis of its 60 years of history.

Crit Rev Oncol Hematol. 2012;84(2):181-199. doi:10.1016/j.critrevonc.2012.03.002.

5. Goodin S. Oral chemotherapeutic agents: Understanding mechanisms of action and drug interactions. Am

J Heal Syst Pharm. 2007;64(9_Supplement_5):S15-S24. doi:10.2146/ajhp070034.

6. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338-345. doi:10.1038/nature12625.

7. Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: Treating cancer with specificity. Eur J Pharmacol. 2018;834:188-196. doi:10.1016/j.ejphar.2018.07.034.

8. Joo WD, Visintin I, Mor G. Targeted cancer therapy-are the days of systemic chemotherapy numbered?

Maturitas. 2013;76(4):308-314. doi:10.1016/j.maturitas.2013.09.008.

9. Lee DH. Treatments for EGFR-mutant non-small cell lung cancer (NSCLC): The road to a success, paved with failures. Pharmacol Ther. 2017;174:1-21. doi:10.1016/j.pharmthera.2017.02.001.

10. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344(11):783-792. doi:10.1056/ NEJM200103153441101.

11. Harbeck N, Gnant M. Breast cancer. Lancet. 2017;389(10074):1134-1150. doi:10.1016/S0140-6736(16)31891-8.

12. Cancer Genome Atlas Research Network. The Cancer Genome Atlas pan-cancer analysis project. Nat Genet. 2013;45(10):1113-1120. doi:10.1038/ng.2764.

13. Barrett T, Wilhite SE, Ledoux P, et al. NCBI GEO: Archive for functional genomics data sets - Update. Nucleic

Acids Res. 2013;41(D1):991-995. doi:10.1093/nar/gks1193.

14. Benstead-Hume G, Wooller SK, Pearl FMG. “Big data” approaches for novel anti-cancer drug discovery.

Expert Opin Drug Discov. 2017;12(6):599-609. doi:10.1080/17460441.2017.1319356.

15. Collisson EA, Campbell JD, Brooks AN, et al. Comprehensive molecular profiling of lung adenocarcinoma.

Nature. 2014;511(7511):543-550. doi:10.1038/nature13385.

16. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology. Non-small cell lung cancer. November 6, 2019.

17. Bell D, Berchuck A, Birrer M, et al. Integrated genomic analyses of ovarian carcinoma. Nature.

1

14 Chapter 1

(8)

2011;474(7353):609-615. doi:10.1038/nature10166.

18. Morgan RD, Clamp AR, Evans DGR, Edmondson RJ, Jayson GC. PARP inhibitors in platinum-sensitive high-grade serous ovarian cancer. Cancer Chemother Pharmacol. 2018;81(4):647-658. doi:10.1007/s00280-018-3532-9.

19. Kuwano M, Sonoda K, Murakami Y, Watari K, Ono M. Overcoming drug resistance to receptor tyrosine kinase inhibitors: Learning from lung cancer. Pharmacol Ther. 2016;161:97-110. doi:10.1016/j. pharmthera.2016.03.002.

20. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr Opin Immunol. 2014;27:16-25. doi:10.1016/j. coi.2014.01.004.

21. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer

Discov. 2018;8(9):1069-1086. doi:10.1158/2159-8290.CD-18-0367.

22. Nadal E, Massuti B, Dómine M, García-Campelo R, Cobo M, Felip E. Immunotherapy with checkpoint inhibitors in non-small cell lung cancer: insights from long-term survivors. Cancer Immunol Immunother. 2019;68(3):341-352. doi:10.1007/s00262-019-02310-2.

23. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small cell lung cancer. N Engl J Med. 2015;372(21):2018-2028. doi:10.1056/NEJMoa1501824.

24. Tobin NP, Foukakis T, De Petris L, Bergh J. The importance of molecular markers for diagnosis and selection of targeted treatments in patients with cancer. J Intern Med. 2015;278(6):545-570. doi:10.1111/joim.12429. 25. Festino L, Botti G, Lorigan P, et al. Cancer treatment with anti-PD-1/PD-L1 agents: is PD-L1 expression a

biomarker for patient selection? Drugs. 2016;76(9):925-945. doi:10.1007/s40265-016-0588-x.

26. Mansfield AS, Aubry MC, Moser JC, et al. Temporal and spatial discordance of programmed cell death-ligand 1 expression and lymphocyte tumor infiltration between paired primary lesions and brain metastases in lung cancer. Ann Oncol. 2016;27(1):1953-1958. doi:10.1093/annonc/mdw289.

27. Bensch F, van der Veen EL, Lub-de Hooge MN, et al. 89Zr-atezolizumab imaging as a non-invasive approach

to assess clinical response to PD-L1 blockade in cancer. Nat Med. 2018;24(12):1852-1858. doi:10.1038/ s41591-018-0255-8.

28. Xin Yu J, Hubbard-Lucey VM, Tang J. The global pipeline of cell therapies for cancer. Nat Rev Drug Discov. May 2019. doi:10.1038/d41573-019-00090-z.

(9)

Referenties

GERELATEERDE DOCUMENTEN

Identification and modulation of drug targets for precision medicine in breast, lung and ovarian cancer subtypes.. University

Abbreviations; TNBC: triple negative breast cancer; HER2: human epidermal growth factor receptor 2; NK: natural killer cell; T reg : regulatory T cell; NKT: Natural killer T cell;

Subsequently, we demonstrate the importance of MAPK signaling in the upregulation of PD-L1 by growth factors and IFNγ in lung adenocarcinoma cell lines, which was mediated

In vitro treatment of the strongly PD-L1 positive ES2 cells with selumetinib for 24 hours resulted in a complete blockade of MEK1/2 signaling and ~50% downregulation of

The research presented in this thesis aimed to provide a biological rationale for potential combination therapies and patient selection tools to enhance efficacy of immunotherapy and

Deze studie toont de kracht van c-ICA bij het ontleden van genexpressiedatasets en onthulde nieuwe biologische processen die mogelijk relevant zijn voor de klinische uitkomsten

aanwezig was wil ik graag bedanken voor hun kritische vragen en alle input voor mijn projecten.. Graag wil ik ook

Identification and modulation of drug targets for precision medicine in breast, lung and ovarian cancer subtypes..