• No results found

University of Groningen Tumor immunology in ovarian cancer Merkus-Brunekreeft, Kim

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Tumor immunology in ovarian cancer Merkus-Brunekreeft, Kim"

Copied!
7
0
0

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

Hele tekst

(1)

University of Groningen

Tumor immunology in ovarian cancer

Merkus-Brunekreeft, Kim

DOI:

10.33612/diss.147014180

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):

Merkus-Brunekreeft, K. (2020). Tumor immunology in ovarian cancer. University of Groningen.

https://doi.org/10.33612/diss.147014180

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)

1

GENERAL INTRODUCTION, AIM,

(3)

13 12

General introduction, aim, and outline

1

GENERAL INTRODUCTION

High-grade serous ovarian cancer

Ovarian cancer is the third most common gynecological malignancy worldwide with an overall 5-year survival of ~35%. As such, ovarian cancer is the deadliest gynecological malignancy and the fifth leading cause of cancer death in women.1,2 In 2018 an

estimated 300,000 new patients were diagnosed with ovarian cancer and in the same year there were an estimated 184,000 ovarian cancer deaths worldwide.3 This poor

prognosis is largely due to diagnosis at an advanced stage, high recurrence rate and of chemo-resistance after initial treatment. Currently there are no adequate screening methods or early detectable symptoms of disease that could lead to earlier diagnosis. Factors that determine long-term survival of ovarian cancer patients are well-known clinicopathological factors, such as stage and surgical outcome, and the presence of tumor-infiltrating lymphocytes (TIL).4 Regretfully, the 5-year overall survival of ovarian

cancer patients has not improved over the past decades.

Ovarian cancer can be divided in various histological subtypes (serous, germ cell tumors, sex cord stromal tumors, mixed cell, clear cell, transitional cell, and undifferentiated) of which epithelial ovarian cancer is the most common subtype. Epithelial ovarian cancers originate either from the epithelial surface of the ovaries or fallopian tubes.5,6

Increasing evidence has shown that the majority of ovarian cancers originates in the fallopian tubes instead of the ovaries themselves.8,9 Serous ovarian cancer is the most

common subtype of epithelial ovarian cancers.10 The studies in this thesis focus on

the most common and aggressive subtype of all: high-grade serous ovarian cancer (HGSOC).

Tumor immunology

Since the beginning of twentieth century, a cytotoxic approach towards malignancies has been chosen over an immune-based strategy, but until today the cytotoxic approach fails to cure late-stage or recurring malignancies, among which is recurring ovarian cancer, notorious for its chemotherapy resistance.11-13 The first clue of immunity

in cancer stems from the late 19th century. In 1893, a surgeon named William B.

Coley developed the first immune-based treatment for cancer. In 1909 the physician-scientist Paul Ehrlich introduced the cancer immunosurveillance hypothesis.14 It took

another century before Coley’s results were connected to Ehrlich’s hypothesis. In 1959,

Ruth and John Graham published the first ever immune-modulating vaccine study in gynecologic cancer, 114 gynecologic cancer patients were treated with autologous tumor lysate, 22% of the patients showed either stable disease or remission of disease, but effects were unpredictable and the mechanism was not understood.15,16

Around the turn of the last century, Schreiber described the phenomenon of cancer immunoediting, describing the relationship between the immune system and tumor cells.17 Catalyzing progressive interest in tumor immunology, which led to the discovery

of checkpoint blockade therapy mediated through programmed cell death protein 1 (PD-L1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and the development of adoptive cell transfer (ACT) therapy.18,19 Immune checkpoint therapy attenuates

tumor microenvironment-induced suppression of T cell receptor (TCR) signaling, thereby re-activating tumor-infiltrating T cells, resulting in cancer cell elimination. The response is generally thought to involve recognition of mutation-induced amino-acid changes, so-called neo-antigens. Clinically, immune checkpoint blockade therapy has proven highly effective in treatment of a number of cancer types, including lung cancer, melanoma and uterine cancer. In ovarian cancer, responses remain limited however. Nevertheless, patients that respond show long-term disease-free survival over periods of years. No surprise, the editors of the leading scientific journal Science announced cancer immunotherapy the breakthrough of the year 2013 and the work of James P. Allison and Tasuku Honjo was honored with the Nobel prize for physiology in 2018.20

Tumor microenvironment

In 1975 Beatrice Mintz and Karl Ilmensee discovered that cancer cells only grow under certain conditions, emphasizing the importance of the tumor microenvironment (TME).21 We and others demonstrated the importance of immune cell infiltration into

the TME of ovarian tumors.22-24 In particular the number of CD8+ T cells is associated

with prolonged survival in patients treated with surgery, but not in patients treated with neoadjuvant chemotherapy.22 The underlying mechanism for this difference

remains unclear, but may be a consequence of changes in antigen-presentation (see also below). In addition, T cells were of prognostic benefit only when intraepithelially located.25 We further investigated this survival benefit in HGSOC patients and

demonstrated that the prognostic effect was not only depending on the number of infiltrating TIL and their location, but also on the treatment regimen, surgical outcome and T cell differentiation status.22 In our work, programmed cell dead ligand

(4)

tumor-1

infiltrating myeloid cells (TIM), such as tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC). MDSC are associated with worse survival in ovarian cancer, at least in part by suppressing CD8+ TIL.26,27

Myeloid cells are temporarily suppressed during chemotherapy and numbers are lower than before and after the chemotherapy. Therefore, this time window could be interesting for T cell augmenting immunotherapies.

Tumor antigen expression and the lymph node micro-environment

For an adequate immune response antigen presentation is necessary. Major histocompatibility complex class 1 (MHC-I) is expressed on the cell surface of all nucleated cells, including immune and tumor cells, and present fragments of intracellular proteins to immune cells. MHC-I expression on myeloid and lymphoid cells is frequently higher than on cancer cells, suggesting an important role in immune regulation.22 Antigen presentation is most effectively done in the lymph nodes and is

essential for activating a T cell immune response.

Tumor draining lymph nodes (tDLN) are under direct influence of tumor-derived factors and are of great importance in establishing an effective immune response by activating naïve immune cells.28,29 For the gynecological malignancies endometrial and

cervical cancer, it was revealed that the draining lymph node immune cell composition is characterized by either an immunosuppressive or naïve signature.30,31 Currently

there is limited data on the immune cell composition of lymph nodes in ovarian cancer. Expanding our knowledge on the tDLN microenvironment in ovarian cancer patients might help us to understand the minimal responses to immunotherapy observed in ovarian cancer so far.

Carcinogenesis in ovarian cancer

In the late 19th century the idea of cancer as genetic disease arose, following several

new genetic-based hypotheses including the Knudson hypothesis, better known as the two-hit model, and the presence of tumor suppressor and oncogenes.32,33 HGSOC

is known for its high level of chromosomal instability, rapid tumor development, and the expression of the typically mutated TP53-gene.34

The lifetime risk of developing ovarian cancer is greater in women who are carriers of the BRCA1, BRCA2, or Lynch Syndrome (LS) mutation. About 1-2% of the general population will develop ovarian cancer, but in case of BRCA1, BRCA2, LS mutation, and TP53 it is respectively 45-55%, 20-30%, 10-15%, and 3%. Known carriers of these genes can intensify screening or undergo risk-reducing surgery, no other preventative

measures are known yet.35

In general, these handful of conserved mutation-induced neo-antigens in ovarian cancer is presumed to be insufficient to effectively target ovarian cancers for immune-mediated destruction. Therefore, methods are developed to identify tumor-specific targets, e.g. antigens, unique to every individual tumor. It is challenging to find tumor-specific antigens that are highly expressed by the tumor cells, but virtually absent in normal tissue, and ideally these antigens are crucial for cancer cell growth to decrease the risk of immune escape due to downregulation of these tumor-specific antigens.36 Therefore, methods are investigated and in part described in this thesis to

identify tumor-specific mutations and target these to improve the tumor-specificity of immunotherapies. These neoantigens can be analyzed for response in preclinical models prior to clinical development.

Preclinical in vivo ovarian cancer models

Existing preclinical in vivo models for ovarian cancer are of limited usage because they are either fully murine, lack a human immune system, or hindered by graft-versus-host-disease (GVHD).37,38 Therefore, a preclinical in vivo model is desired that constitutes

the human tumor and immune system but is not hampered by GVHD, however such model is hard to establish.

One of the many investigated models is the patient-derived xenograft (PDX) model. Ovarian cancer (OC) PDX models are valuable since they largely preserve the complexity of the OC tumor microenvironment.39 OC PDX models can be established in various

ways; however, all of these models lack a functional human immune system. Various methods are investigated to introduce components of the human immune system into PDX models: (1) the engraftment of donor-unrelated CD34 human hematopoietic stem cells and (2) the injection of human immune cells.40-44 Although the first method

introduces both the innate and adaptive immune system in the murine model and is not hampered by the development of GVHD, it is of limited value because of the lack of thymic education of the T cells. The second model reconstitutes (partial) the immune system in the PDX models but is limited by the early onset of GVHD, ethical restrictions and only partial reconstitution of the human immune system in mice. A solution for this problem could be the use of donor-matched immune cells, preferably tumor-specific immune cells.45

Another challenge is the limited access to and availability of patient material. This poses several other challenges for the establishment of a sustainable preclinical OC PDX model since large quantities (>1x108 cells) of immune cells are needed to obtain

(5)

17 16

General introduction, aim, and outline

1

clinical results and numerous immune-humanized mice are necessary for statistically significant results.46 Therefore, methods are sought to generate more tumor tissue,

cells and ways to identify and select tumor-specific T cells, and (clonally) expand these cells for therapy. In 2018 it was shown that it is possible to identify neo-antigen specific T cells, even in low mutational tumors like ovarian cancer, paving the way for mutation-based personal therapy. However, they were unable to establish a preclinical in vivo model to validate their findings.47 In 2019 it was demonstrated

that neoantigens found in a primary ovarian tumor were retained in a patient-derived ovarian cancer model and the possible activation of autologous T cells due to these neoantigens.48

Until a proper preclinical in vivo model is established an alternative could be using ovarian cancer (OC) organoids. Köpper et al. (2019) managed to establish 56 OC organoids of 32 patients that can be used for drug-screening assays and maintain the histological and genomic characteristics of the original tumor.49 Even more importantly,

the organoids can be xenografted in murine models enabling preclinical in vivo testing of treatments demonstrating their potential usage in the field of personalized medicine.

AIM OF THIS THESIS

Increasing the understanding of the role of the immune system in ovarian cancer in relation to the current standard-of-care treatments and exploring possible new treatment strategies for patients with ovarian cancer.

(6)

1

OUTLINE OF THIS THESIS

Chapter 2 demonstrates that immune contexture is defined by tissue of origin and

independent of chemotherapy. It is shown that draining lymph nodes in ovarian cancer patients are characterized by a quiescent microenvironment and that T cell differentiation is heterogeneous across tumor tissue, lymph nodes and in the peripheral blood.

Chapter 3 shows that CD103+ TIL in HGSOC are phenotypically diverse TCR TCRαβ+

CD8αβ+ T cells expressing the therapeutic targets PD-1 and CD27, indicating that

CD103+ TIL are formed as a result of an active antitumor response that possibly can

be reactivated by checkpoint inhibition.

Chapter 4 shows that minimal major histocompatibility complex class 1 (MHC-I)

expression after neoadjuvant chemotherapy in ovarian cancer is negatively associated with T-cell-dependent outcome. This may explain why T cell infiltration in tumors of chemotherapy-naïve patients is associated with a favorable prognosis and not in tissue of patients that received chemotherapy prior to surgery.

Chapter 5 discusses the construction and in vitro validation of fusion-proteins

specifically delivering CD40 ligand (CD40L) to various types of malignant cells, including ovarian cancer, showing immune cell activation, proliferation and tumor cell apoptosis. This strategy acknowledges the importance of proper antigen presentation to surveying T cells.

Chapter 6 describes the establishment of an ovarian cancer patient-derived xenograft

model and our view how to introduce human immune system components to the model. Furthermore, it discusses the identification of tumor-specific neoantigens and the process of selecting neoantigen specific T cells and expanding them.

In chapter 7, the significance of the abovementioned studies is summarized. Followed by a general discussion, conclusion and future perspectives.

In chapter 8 a Dutch summary for layman is found. In the appendices, a list of abbreviations; list of contributing authors; list of publications; acknowledgements and curriculum vitae of the author of this PhD-thesis can be found.

REFERENCES

1. Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin 2018 Jul;68(4):284-296.

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018 Jan;68(1):7-30. 3. 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 Nov;68(6):394-424.

4. Wouters MC, Komdeur FL, de Bruyn M, Nijman HW. Size matters: Survival benefit conferred by intratumoral T cells is dependent on surgical outcome, treatment sequence and T cell differentiation. Oncoimmunology 2016 Jan 4;5(5):e1122863.

5. Piek JM, Dorsman JC, Zweemer RP, Verheijen RH, van Diest PJ, Colgan TJ. Women harboring BRCA1/2 germline mutations are at risk for breast and female adnexal carcinoma. Int J Gynecol Pathol 2003 Jul;22(3):315-6; author reply 315-6.

6. George SH, Garcia R, Slomovitz BM. Ovarian Cancer: The Fallopian Tube as the Site of Origin and Opportunities for Prevention. Front Oncol 2016 May 2;6:108.

7. Daly MB, Dresher CW, Yates MS, Jeter JM, Karlan BY, Alberts DS, et al. Salpingectomy as a means to reduce ovarian cancer risk. Cancer Prev Res (Phila) 2015 May;8(5):342-348.

8. Balsarkar G. Opportunistic Salpingectomy as an Ovarian Cancer Primary Prevention Strategy. J Obstet Gynaecol India 2017 Aug;67(4):243-246.

9. Kim M, Kim YH, Kim YB, Kim J, Kim JW, Park MH, et al. Bilateral salpingectomy to reduce the risk of ovarian/fallopian/peritoneal cancer in women at average risk: a position statement of the Korean Society of Obstetrics and Gynecology (KSOG). Obstet Gynecol Sci 2018 Sep;61(5):542-552. 10. Vaughan S, Coward JI, Bast RC,Jr, Berchuck A, Berek JS, Brenton JD, et al. Rethinking ovarian

cancer: recommendations for improving outcomes. Nat Rev Cancer 2011 Sep 23;11(10):719-725. 11. Friedenreich CM, Shaw E, Neilson HK, Brenner DR. Epidemiology and biology of physical activity

and cancer recurrence. J Mol Med (Berl) 2017 Oct;95(10):1029-1041.

12. Eisenhauer EA, Vermorken JB, van Glabbeke M. Predictors of response to subsequent chemotherapy in platinum pretreated ovarian cancer: a multivariate analysis of 704 patients [seecomments. Ann Oncol 1997 Oct;8(10):963-968.

13. Markman M, Rothman R, Hakes T, Reichman B, Hoskins W, Rubin S, et al. Second-line platinum therapy in patients with ovarian cancer previously treated with cisplatin. J Clin Oncol 1991 Mar;9(3):389-393.

14. Ehrlich O. Ueber den jetzigen Stand der Karzinomforschung. Ned Tijdschr Geneeskd.

1909;53:273-90

15. Graham JB, Graham RM. The effect of vaccine on cancer patients. Surg Gynecol Obstet 1959 Aug;109(2):131-138.

16. Decker WK, da Silva RF, Sanabria MH, Angelo LS, Guimaraes F, Burt BM, et al. Cancer Immunotherapy: Historical Perspective of a Clinical Revolution and Emerging Preclinical Animal Models. Front Immunol 2017 Aug 2;8:829.

(7)

21 20

General introduction, aim, and outline

1

17. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity 2004 Aug;21(2):137-148.

18. MITCHISON NA. Studies on the immunological response to foreign tumor transplants in the mouse. I. The role of lymph node cells in conferring immunity by adoptive transfer. J Exp Med 1955 Aug 1;102(2):157-177.

19. Fan J, Shang D, Han B, Song J, Chen H, Yang JM. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Theranostics 2018 Nov 10;8(20):5784-5800.

20. Guo ZS. The 2018 Nobel Prize in medicine goes to cancer immunotherapy (editorial for BMC cancer). BMC Cancer 2018 Nov 12;18(1):1086-018-5020-3.

21. Mintz B, Illmensee K. Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc Natl Acad Sci U S A 1975 Sep;72(9):3585-3589.

22. Wouters MC, Komdeur FL, Workel HH, Klip HG, Plat A, Kooi NM, et al. Treatment Regimen, Surgical Outcome, and T-cell Differentiation Influence Prognostic Benefit of Tumor-Infiltrating Lymphocytes in High-Grade Serous Ovarian Cancer. Clin Cancer Res 2016 Feb 1;22(3):714-724.

23. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. Intraepithelial CD8 tumor-infiltrating lymphocytes and a high CD8/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A 2005 Dec 20;102(51):18538-18543.

24. Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol 2012 Feb;124(2):192-198. 25. Workel HH, Komdeur FL, Wouters MC, Plat A, Klip HG, Eggink FA, et al. CD103 defines intraepithelial

CD8 PD1 tumour-infiltrating lymphocytes of prognostic significance in endometrial adenocarcinoma. Eur J Cancer 2016 Jun;60:1-11.

26. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012 Mar 22;12(4):253-268.

27. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 2010 Oct;11(10):889-896.

28. Munn DH, Mellor AL. The tumor-draining lymph node as an immune-privileged site. Immunol Rev 2006 Oct;213:146-158.

29. Cochran AJ, Huang RR, Lee J, Itakura E, Leong SP, Essner R. Tumour-induced immune modulation of sentinel lymph nodes. Nat Rev Immunol 2006 Sep;6(9):659-670.

30. Heeren AM, Koster BD, Samuels S, Ferns DM, Chondronasiou D, Kenter GG, et al. High and interrelated rates of PD-L1+CD14+ antigen-presenting cells and regulatory T cells mark the

microenvironment of metastatic lymph nodes from patients with cervical cancer. Cancer Immunol Res 2015 Jan;3(1):48-58.

31. Fattorossi A, Battaglia A, Ferrandina G, Buzzonetti A, Legge F, Salutari V, et al. Lymphocyte composition of tumor draining lymph nodes from cervical and endometrial cancer patients. Gynecol Oncol 2004 Jan;92(1):106-115.

32. Knudson AG,Jr. Hereditary cancer, oncogenes, and antioncogenes. Cancer Res 1985 Apr;45(4):1437-1443.

33. Knudson AG. Hereditary cancer: two hits revisited. J Cancer Res Clin Oncol 1996;122(3):135-140.

34. Vang R, Shih I, Kurman RJ. Ovarian low-grade and high-grade serous carcinoma: pathogenesis, clinicopathologic and molecular biologic features, and diagnostic problems. Adv Anat Pathol 2009 Sep;16(5):267-282.

35. Toss A, Tomasello C, Razzaboni E, Contu G, Grandi G, Cagnacci A, et al. Hereditary ovarian cancer: not only BRCA 1 and 2 genes. Biomed Res Int 2015;2015:341723.

36. Miller AM, Bahmanof M, Zehn D, Cohen EEW, Schoenberger SP. Leveraging TCR Affinity in Adoptive Immunotherapy against Shared Tumor/Self-Antigens. Cancer Immunol Res 2019 Jan;7(1):40-49. 37. Magnotti E, Marasco WA. The latest animal models of ovarian cancer for novel drug discovery.

Expert Opin Drug Discov 2018 Mar;13(3):249-257.

38. King MA, Covassin L, Brehm MA, Racki W, Pearson T, Leif J, et al. Human peripheral blood leucocyte non-obese diabetic-severe combined immunodeficiency interleukin-2 receptor gamma chain gene mouse model of xenogeneic graft-versus-host-like disease and the role of host major histocompatibility complex. Clin Exp Immunol 2009 Jul;157(1):104-118.

39. Alkema NG, Tomar T, Duiker EW, Jan Meersma G, Klip H, van der Zee AG, et al. Biobanking of patient and patient-derived xenograft ovarian tumour tissue: efficient preservation with low and high fetal calf serum based methods. Sci Rep 2015 Oct 6;5:14495.

40. Bankert RB, Balu-Iyer SV, Odunsi K, Shultz LD, Kelleher RJ,Jr, Barnas JL, et al. Humanized mouse model of ovarian cancer recapitulates patient solid tumor progression, ascites formation, and metastasis. PLoS One 2011;6(9):e24420.

41. Koneru M, Purdon TJ, Spriggs D, Koneru S, Brentjens RJ. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology 2015 Jan 23;4(3):e994446.

42. Chang DK, Peterson E, Sun J, Goudie C, Drapkin RI, Liu JF, et al. Anti-CCR4 monoclonal antibody enhances antitumor immunity by modulating tumor-infiltrating Tregs in an ovarian cancer xenograft humanized mouse model. Oncoimmunology 2015 Dec 10;5(3):e1090075.

43. Wang W, Kryczek I, Dostal L, Lin H, Tan L, Zhao L, et al. Effector T Cells Abrogate Stroma-Mediated Chemoresistance in Ovarian Cancer. Cell 2016 May 19;165(5):1092-1105.

44. Heo EJ, Cho YJ, Cho WC, Hong JE, Jeon HK, Oh DY, et al. Patient-Derived Xenograft Models of Epithelial Ovarian Cancer for Preclinical Studies. Cancer Res Treat 2017 Oct;49(4):915-926. 45. Yong KSM, Her Z, Chen Q. Humanized Mice as Unique Tools for Human-Specific Studies.

Arch Immunol Ther Exp (Warsz) 2018 Aug;66(4):245-266.

46. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J Immunother 2003 Jul-Aug;26(4):332-342.

47. Bobisse S, Genolet R, Roberti A, Tanyi JL, Racle J, Stevenson BJ, et al. Sensitive and frequent identification of high avidity neo-epitope specific CD8 () T cells in immunotherapy-naive ovarian cancer. Nat Commun 2018 Mar 15;9(1):1092-018-03301-0.

48. Want MY, Konstorum A, Huang RY, Jain V, Matsueda S, Tsuji T, et al. Neoantigens retention in patient derived xenograft models mediates autologous T cells activation in ovarian cancer. Oncoimmunology 2019 Mar 30;8(6):e1586042.

49. Kopper O, de Witte CJ, Lohmussaar K, Valle-Inclan JE, Hami N, Kester L, et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med 2019 May;25(5):838-849

Referenties

GERELATEERDE DOCUMENTEN

On the contrary, when examining this effect in a larger cohort by comparing the expression of EpCAM in malignant tissues from patients who underwent primary cytoreductive surgery

as GSK126 only inhibit cell growth of ARID1A mutated cells and not wild type, because EZH2 inhibition only affects PIK3IP1 expression when ARID1A is

Paraffin embedded tonsil tissue was used as a positive control for quadruple staining of different T-cell subsets containing different markers: CD3, CD27 and CD8 (DAPI was used

This thesis consists of fundamental studies on the immune cell populations in various immune cell compartments of ovarian cancer patients: the tumor, the peripheral blood,

CD27 op geactiveerde maar geremde afweercellen; de afwezigheid van MHC-I op tumorcellen die aan chemotherapie zijn blootgesteld; het gebruik van fusie-eiwitten om het afweersysteem

University Medical Center Groningen Department of Obstetrics and Gynecology Groningen, The

A lack of T cell activation in ovarian cancer tumor draining lymph nodes might explain the relatively poor response of ovarian cancer patients to current

2010, "Circulating tumor cells with a putative stem cell phenotype in peripheral blood of patients with breast cancer", Cancer letters, vol. &