• No results found

University of Groningen Preclinical evaluation and molecular imaging of HER family dynamics to guide cancer therapy Kol, Klaas Jan-Derk

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Preclinical evaluation and molecular imaging of HER family dynamics to guide cancer therapy Kol, Klaas Jan-Derk"

Copied!
11
0
0

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

Hele tekst

(1)

Preclinical evaluation and molecular imaging of HER family dynamics to guide cancer therapy

Kol, Klaas Jan-Derk

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: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Kol, K. J-D. (2019). Preclinical evaluation and molecular imaging of HER family dynamics to guide cancer therapy. Rijksuniversiteit Groningen.

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)

CHAPTER

(3)

BACKGROUND

Cancer remains one of the leading causes of death worldwide (1). Current cancer treatment strategies consist of surgery, radiotherapy and systemic treatment. Using conventional multimodality therapy, the survival rates for certain tumor types still could benefit from major improvements (2). Moreover, the cancer incidence is increasing globally. This demonstrates an urgent need for new, effective anti-cancer therapies. During the past two decades, the paradigm for cancer treatment has expanded alongside relatively nonspecific chemotherapeutic drugs, such as taxanes and platinum-based anticancer drugs, to molecularly targeted therapeutics and cancer immunotherapy. Targeted therapies aim to inhibit molecular pathways important for tumor growth and survival, whereas cancer immunotherapy stimulates a host immune response to induce tumor cell kill. Targeted therapies can also have an effect on the immune system by affecting pathways that are crucial for immune development and function. Moreover, therapeutic monoclonal antibodies targeting specific tumor antigens can induce antibody-dependent cellular cytotoxicity (ADCC). Combining these approaches are part of current treatment strategies for many types of cancer, including lung, breast, colon, gastric, head and neck and ovarian cancer as well as melanoma.

One of the first known molecular targets has been the extensively studied human epidermal growth factor receptor (HER) family of receptor tyrosine kinases. The family comprising the epidermal growth factor receptor (EGFR) (also known as HER1), HER2, HER3 and HER4, is not only essential for the development and maintenance of normal tissues, but is also strongly involved in the development of many types of cancer (3,4). Upon ligand binding, these receptors homodimerize or interact with each other, forming heterodimers. Dimerization of the receptors results in activation of downstream signaling cascades such as the RAS-extracellular signal-regulated kinase (ERK) pathway and phosphoinositide 3-kinase (PI3K)/Akt pathway, thereby controlling many biological processes in normal tissue. However, these HER signaling pathways are frequently hyperactivated in cancer due to receptor overexpression, autocrine stimulation, crosstalk with other receptors and/or mutations in components of these pathways (5).

EGFR and HER2 are widely used targets for cancer therapy. In view of its overexpression in tumors and compensatory role in HER signaling, HER3 has gained much interest as an additional potential drug target as well. Drugs that block HER family members or inhibit downstream signaling by HER family members, including human anti-HER antibodies and tyrosine kinase inhibitors (TKIs), are increasingly becoming available. Anti-HER monoclonal antibodies can exert their action via a variety of mechanisms, including blocking (hetero)dimerization and ligand binding, as well as inducing HER endocytosis, complement dependent cytotoxicity (CDC) and ADCC (6–10). Several EGFR- and

(4)

HER2-11

General introduction

1

targeted monoclonal antibodies and TKIs have already been approved by the Food and Drug Administration USA (11). Furthermore, a number of monoclonal antibodies that target HER3 have entered clinical trials (12). Nonetheless, intrinsic and acquired resistance against HER-targeted agents is a clinical problem, and much research focuses on optimizing targeting of the HER family (13,14).

Rapid non-invasive assessment of changes in HER protein expression as a result of treatment with HER-targeted agents would be of major interest for clinical decision making. HER-targeted agents affect many processes involved in HER protein expression, dynamics and availability, such as a) ligand binding, b) receptor dimerization, shedding, internalization, recycling to the cell surface and degradation, c) mobilization of internal receptor pools, and d) induction of HER family gene expression. In addition, the size of receptor-antibody complexes influences the amount of internalization and recycling, and subsequent degradation of receptors (15). A good example of these drug-induced changes in HER protein dynamics is the treatment of HER2-driven breast cancer with the EGFR/HER2 TKI lapatinib that has been shown to induce a transcriptional and posttranslational compensatory increase in HER3, which may attenuate antitumor efficacy of this drug (16).

Because of the dynamic HER membrane expression, a non-invasive dynamic assessment of HER tumor status in vivo will provide important supplementary information that cannot be obtained with invasive static techniques such as immunohistochemistry on biopsy material. Non-invasive molecular imaging, defined as the in vivo characterization and measurement of biological processes at the cellular and molecular level, could potentially be used as a tool to monitor receptor dynamics (17). Molecular characteristics, such as HER cell surface expression, can be imaged with three-dimensional whole body positron emission tomography (PET) using antibodies radiolabeled with zirconium-89 (89Zr, t½ = 78.4 h), for optimal lesion accumulation. HER antibody-based PET-imaging

could play an important role in identifying patients who might benefit from anti-HER antibody therapy, as well as monitoring responses to HER-targeting agents. Several (pre-) clinical studies have assessed in vivo treatment effects of drugs targeting HER2 such as lapatinib, trastuzumab or HSP90 inhibitors by quantifying the (change in) radiolabeled tracer accumulation in tumors (18–22). Results showed that these drugs reduced tumor uptake of radiolabeled anti-HER2 antibodies. HER-targeted therapies can also influence dynamics of other plasma membrane proteins, such as immune checkpoints. Immune checkpoints refer to multiple co-stimulatory or inhibitory protein interactions that (among others) regulate T cell responses to antigens (23,24). These protein interactions are essential in self-tolerance and limit collateral

(5)

tissue damage when the immune system is responding to pathogenic infections. Tumors can exploit these immune checkpoints by upregulating inhibitory proteins and downregulating stimulatory proteins. This dampens T cell effector functions by inhibiting signaling downstream of the T cell receptor and, as a consequence, protects tumors from immune-mediated rejection. Programmed cell death 1 (PD-1) and its ligand PD-L1 have emerged as critical immune checkpoint proteins in cancer. PD-L1 expressed on tumor cells causes anergy and apoptosis of activated T cells by binding to PD-1 expressed on the surface of these T cells. PD-1/PD-L1 checkpoint blockade with antibodies, which are examples of cancer immunotherapy, has resulted in significant long lasting responses in patients with advanced-stage cancers, including heavily pretreated non-small cell lung cancer (NSCLC) and melanoma patients (25–27). Recently, it has been shown that EGFR plays a role in PD-L1 expression by tumor cells. EGFR inhibition reduces PD-L1 expression

in vitro, resulting in a concomitant increase in T cell activation (28,29). Therefore, EGFR

inhibition may be used to improve efficacy of immunotherapy.

Taken together, these results indicate that plasma membrane protein dynamics might well be important for proper design of (combination) therapies. Therefore, the aim of this thesis is to gain more insight in the effect of HER-targeting agents on HER and PD-L1 dynamics to provide a rationale for future combination therapies. In addition, HER tracers for molecular imaging to evaluate drug tumor targeting, organ distribution and target dynamics are explored.

OUTLINE OF THESIS

Anti-EGFR monoclonal antibody combinations can effectively inhibit the EGFR signaling pathway, and have shown superior anticancer efficacy in several human tumor xenograft models (30–32). Cetuximab and imgatuzumab are both monoclonal antibodies directed against distinct, non-overlapping epitopes in EGFR extracellular domain III. Imgatuzumab, in addition, is glycoengineered to enhance ADCC responses (33). Thus, combining these antibodies is a potential strategy to target EGFR more effectively than existing treatments with single antibodies. It is unknown whether treatment with imgatuzumab or the combination with cetuximab increases EGFR internalization and/or receptor membrane recycling in cancer cells, potentially reducing ADCC responses. In chapter 2, we investigate the effects of imgatuzumab and cetuximab on EGFR dynamics and

intracellular signaling. In addition, we monitor whether changes in EGFR dynamics affect ADCC responses and tumor cell growth inhibition. Effects of imgatuzumab, cetuximab and the combination of these monoclonal antibodies on EGFR are studied in a panel of NSCLC cell lines. The effect of antibody binding on EGFR cell surface dynamics is determined using flow cytometry and immunofluorescence. In addition, the effect of antibody binding on total EGFR protein levels, downstream signaling and growth are

(6)

13

General introduction

1

studied using Western blotting and proliferation assays. Finally, ADCC assays are used to investigate effects of EGFR dynamics on ADCC responses.

Whole body analysis of EGFR expression in tumor lesions using PET imaging could support decision making during clinical development and clinical practice. Preclinical EGFR-microPET imaging revealed a mismatch between in vivo tumor EGFR expression assessed using Western blotting and tumor uptake of the radiolabeled EGFR antibody tracer (34). Shed EGFR ectodomain, which is present in cancer patient sera, can potentially bind tracer and therefore affect tracer kinetics (35). In chapter 3, we examine the influence

of shed EGFR levels on tracer kinetics and tumor uptake of EGFR monoclonal antibody

89Zr-imgatuzumab in varying xenograft models in order to optimize 89Zr-imgatuzumab

PET. Human A431 (EGFR overexpressing, epidermoid carcinoma of the vulva), A549 and H441 (both EGFR medium expressing, NSCLC) cancer cell lines are xenografted in mice. The A431 cell line is used, because of its known EGFR shedding potential. Tumor bearing mice receive various doses of 89Zr-imgatuzumab, co-injected with equal doses

of 111In-IgG as internal control. MicroPET scans are performed at multiple time points

post tracer injection, followed by biodistribution analysis. Shed EGFR levels in liver and plasma samples, as well as in vitro culture media are determined by ELISA.

HER3 is the only member of the HER family having impaired tyrosine kinase activity and therefore its role in cancer has long been underestimated. However, in view of its overexpression in various tumor types and its compensatory role in HER signaling, HER3 has gained much interest as a potential target in cancer treatment. A review, chapter 4,

describes the biology and relevance of HER3 in cancer, as well as drugs that block HER3 or interfere with HER3 heterodimer signaling. These drugs include fully human anti-HER3 antibodies, bispecific antibodies and TKIs, and are currently becoming available for clinical use. An overview of HER3-targeting drugs in clinical trials is given. Biomarkers might be useful for prediction and monitoring of treatment effects, as well as supporting decision making during clinical development and clinical practice. Therefore, potential biomarkers for effective HER3-targeting such as tumor analysis of HER3 expression, functional assays for downstream effector molecules and molecular imaging techniques are discussed.

HER3 membrane expression is very dynamic due to its compensatory role in HER signaling, particularly in response to drugs targeting HER family members or the PI3K pathway, which may attenuate the antitumor action of these inhibitors (13, 16, 36, 37). Preclinically, combining HER3 with HER2 inhibitors augmented tumor growth inhibition,

in vivo (38). This might advocate for a more dynamic assessment of HER3 tumor status

(7)

status assessment with 89Zr-labeled anti-HER3 antibody mAb3481 PET after treatment

with the dual EGFR/HER2 inhibitor lapatinib. The effect of lapatinib on HER3 cell surface expression and mAb3481 internalization is evaluated in human breast (BT474, SKBR3) and gastric (N87) cancer cell lines using flow cytometry. Next, mice bearing BT474 or N87 xenografts receive lapatinib daily for 9 days. PET-scans of xenografts with 89

Zr-mAb3481 for visualization of HER3 are performed after 9 days lapatinib treatment. PET imaging and ex vivo organ distribution data are compared with Western blot and immunohistochemical stainings of tumor tissue.

Recently, it was shown that constitutively activated mutant EGFR induces PD-L1 expression in NSCLC cell lines (29). EGFR inhibition reduces PD-L1 expression of these EGFR mutant cell lines, resulting in a concomitant increase in T cell activation However, in the patient setting, EGFR mutant NSCLC tumors do not respond to PD-1 blockade, even when PD-L1 expression is high. This is due to several factors such as lack of CD8+ T cell infiltration and low tumor mutational burden (39). In contrast, PD-1 blockade has improved survival of EGFR wild-type NSCLC patients (39–41). Surprisingly, there is only limited data about the regulation of PD-L1 expression in EGFR wild-type NSCLC (42). Improved understanding of PD-L1 regulation may provide a rationale to combine immune checkpoint inhibitors with other targeted agents. Therefore, in chapter 6, we aim to identify pathways that

regulate PD-L1 expression in EGFR wild-type NSCLC by using RNA-sequencing data from The Cancer Genome Atlas (TCGA) lung adenocarcinoma and squamous cell lung carcinoma data sets. We functionally validate our findings using a panel of EGFR wild-type lung adenocarcinoma cell lines and cocultures with peripheral blood mononuclear cells. We assess the effect of the combination interferon gamma (IFNγ), the most potent inducer of PD-L1 expression (43), and EGF, an activator of EGFR-mediated signaling, on PD-L1 mRNA, protein and cell surface expression. Next, we investigate the effect of direct targeting of EGFR with cetuximab and erlotinib, or targeting of downstream signaling with XL147 (PI3K), everolimus (mammalian target of rapamycin 1 (mTORC1) and selumetinib (MAP/ERK kinase 1/2 (MEK1/2)) on PD-L1 expression. Effects on PD-L1 expression and downstream signaling are studied using qPCR, Western blotting and flow cytometry.

In chapter 7, the findings of this thesis are summarized, followed by a general discussion

with future perspectives.

REFERENCES

1. Jemal A, Bray F, Center MM, Ferley J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.

(8)

15

General introduction

1

2002 to 2030. PLoS Med. 2006;3:2011–30.

3. Campbell MR, Amin D, Moasser MM. HER3 comes of age: new insights into its functions and role in signaling, tumor biology, and cancer therapy. Clin Cancer Res. 2010;16:1373–83.

4. Arteaga CL, Engelman JA. ERBB receptors: From oncogene discovery to basic science to mechanism-based cancer therapeutics. Cancer Cell. 2014;25:282–303.

5. Citri A, Yarden Y. ERBB signalling: towards the systems level. Nat Rev Mol Cell Biol. 2006;7:505-16.

6. Li S, Schmitz KR, Jeffrey PD, Wiltzius JJW, Kussie P, Ferguson KM. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell. 2005;7:301–11.

7. Kurai J, Chikumi H, Hashimoto K, Yamaguchi K, Yamasaki A, Sako T, et al. Antibody-dependent cellular cytotoxicity mediated by cetuximab against lung cancer cell lines. Clin Cancer Res. 2007;13:1552–61.

8. Schneider-Merck T, Lammerts van Bueren JJ, Berger S, Rossen K, van Berkel PHC, Derer S, et al. Human IgG2 antibodies against epidermal growth factor receptor effectively trigger antibody-dependent cellular cytotoxicity but, in contrast to IgG1, only by cells of myeloid lineage. J Immunol. 2010;184:512–20.

9. Foon KA, Yang XD, Weiner LM, Belldegrun AS, Figlin RA, Crawford J, et al. Preclinical and clinical evaluations of ABX-EGF, a fully human anti-epidermal growth factor receptor antibody. Int J Radiat Oncol Biol Phys. 2004;58:984–90.

10. Dechant M, Weisner W, Berger S, Peipp M, Beyer T, Schneider-Merck T, et al. Complement-dependent tumor cell lysis triggered by combinations of epidermal growth factor receptor antibodies. Cancer Res. 2008;68:4998–5003.

11. de Gramont A, Watson S, Ellis LM, Rodón J, Tabernero J, de Gramont A, et al. Pragmatic issues in biomarker evaluation for targeted therapies in cancer. Nat Rev Clin Oncol. 2015;12:197–212.

12. Kol A, Terwisscha van Scheltinga AGT, Timmer-Bosscha H, Lamberts LE, Bensch F, de Vries EGE, et al. HER3, serious partner in crime: therapeutic approaches and potential biomarkers for effect of HER3-targeting. Pharmacol Ther. 2014;143:1–11. 13. Sergina NV, Rausch M, Wang D, Blair J, Hann B, Shokat KM, et al. Escape from

HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature. 2007;445:437–41.

14. Garrett JT, Arteaga CL. Resistance to HER2-directed antibodies and tyrosine kinase inhibitors: Mechanisms and clinical implications. Cancer Biol Ther. 2011;11:793– 800.

15. Berger C, Madshus IH, Stang E. Cetuximab in combination with anti-human IgG antibodies efficiently down-regulates the EGF receptor by macropinocytosis. Exp Cell Res. 2012;318:2578–91.

(9)

16. Garrett JT, Olivares MG, Rinehart C, Granja-Ingram ND, Sánchez V, Chakrabarty A, et al. Transcriptional and posttranslational up-regulation of HER3 (ErbB3) compensates for inhibition of the HER2 tyrosine kinase. Proc Natl Acad Sci U S A. 2011;108:5021– 6.

17. Mankoff DA. A definition of molecular imaging. J Nucl Med. 2007 ;48:18N, 21N. 18. McLarty K, Cornelissen B, Cai Z, Scollard DA, Costantini DL, Done SJ, et al.

Micro-SPECT/CT with 111In-DTPA-pertuzumab sensitively detects trastuzumab-mediated

HER2 downregulation and tumor response in athymic mice bearing MDA-MB-361 human breast cancer xenografts. J Nucl Med. 2009;50:1340–8.

19. Oude Munnink TH, de Korte MA, Nagengast WB, Timmer-Bosscha H, Schröder CP, de Jong JR, et al. 89Zr-trastuzumab PET visualises HER2 downregulation by the HSP90

inhibitor NVP-AUY922 in a human tumour xenograft. Eur J Cancer. 2010;46:678–84. 20. Oude Munnink TH, de Vries EGE, Vedelaar SR, Timmer-Bosscha H, Schröder CP, Brouwers AH, Lub-de Hooge MN. Lapatinib and 17AAG reduce 89Zr-trastuzumab-F

(ab’) 2 uptake in SKBR3 tumor xenografts. Mol Pharm. 2012;9:2995-3002.

21. Gaykema SBM, Brouwers AH, Hovenga S, Lub-de Hooge MN, de Vries EGE, Schröder CP. Zirconium-89-trastuzumab positron emission tomography as a tool to solve a clinical dilemma in a patient with breast cancer. J Clin Oncol. 2012;30:e74-5. 22. Gaykema SBM, Schröder CP, Vitfell-Rasmussen J, Chua S, Oude Munnink TH,

Brouwers AH, et al. 89Zr-trastuzumab and 89Zr-bevacizumab PET to evaluate the

effect of the HSP90 inhibitor NVP-AUY922 in metastatic breast cancer patients. Clin Cancer Res. 2014;20:3945–54.

23. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.

24. Smyth MJ, Ngiow SF, Ribas A, Teng MWL. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol. 2015;13:1–16. 25. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E, et al.

Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373:123–35.

26. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti–PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455-65.

27. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122– 33.

28. Chen N, Fang W, Zhan J, Hong S, Tang Y, Kang S, et al. Up-regulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC. J Thorac Oncol. 2015;10:910-23.

(10)

17

General introduction

1

Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 2013;3:1355–63.

30. Spangler JB, Neil JR, Abramovitch S, Yarden Y, White FM, Lauffenburger DA, et al. Combination antibody treatment down-regulates epidermal growth factor receptor by inhibiting endosomal recycling. Proc Natl Acad Sci U S A. 2010;107:13252–7. 31. Pedersen MW, Jacobsen HJ, Koefoed K, Hey A, Pyke C, Haurum JS, et al. Sym004:

a novel synergistic anti-epidermal growth factor receptor antibody mixture with superior anticancer efficacy. Cancer Res. 2010;70:588–97.

32. Ferraro DA, Gaborit N, Maron R, Cohen-Dvashi H, Porat Z, Pareja F, et al. Inhibition of triple-negative breast cancer models by combinations of antibodies to EGFR. Proc Natl Acad Sci U S A. 2013;110:1815–20.

33. Gerdes CA, Nicolini VG, Herter S, van Puijenbroek E, Lang S, Roemmele M, et al. GA201 (RG7160): a novel, humanized, glycoengineered anti-EGFR antibody with enhanced ADCC and superior in vivo efficacy compared with cetuximab. Clin Cancer Res. 2013;19:1126–38.

34. Aerts HJWL, Dubois L, Perk L, Vermaelen P, van Dongen GAMS, Wouters BG, et al. Disparity between in vivo EGFR expression and 89Zr-labeled cetuximab uptake

assessed with PET. J Nucl Med. 2008;50:123–31.

35. Wilken JA, Baron AT, Maihle NJ. The epidermal growth factor receptor conundrum. Cancer. 2011;117:2353–60.

36. Serra V, Scaltriti M, Prudkin L, Eichhorn PJA, Ibrahim YH, Chandarlapaty S, et al. PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer. Oncogene. 2011;30:2547–57.

37. Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19:58–71.

38. Garrett JT, Sutton CR, Kuba MG, Cook RS, Arteaga CL. Dual blockade of HER2 in HER2-overexpressing tumor cells does not completely eliminate HER3 function. Clin Cancer Res. 2013;19:610–9.

39. Dong ZY, Zhang JT, Liu SY, Su J, Zhang C, Xie Z, Zhou Q, Tu HY, Xu CR, Yan LX, et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology. 2017;6.

40. Li J, Chen Y, Shi X, Le X, Feng F, Chen J, Zhou C, Chen Y, Wen S, Zeng H, et al. A systematic and genome-wide correlation meta-analysis of PD-L1 expression and targetable NSCLC driver genes. J Thorac Dis. 2017;9:2560–2571.

41. Zhang M, Li G, Wang Y, Wang Y, Zhao S, Haihong P, Zhao H, Wang Y. PD-L1 expression in lung cancer and its correlation with driver mutations: a meta-analysis. Sci Rep 2017;7:10255.

(11)

42. Lastwika KJ, Wilson W 3rd, Li QK, Norris J, Xu H, Ghazarian SR, et al. Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer. Cancer Res. 2016;76):227-38.

43. Lee SJ, Jang BC, Lee SW, Yang YI, Suh SI, Park YM, et al. Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-gamma-induced upregulation of B7-H1 (CD274). FEBS Lett. 2006;580:755–62.

Referenties

GERELATEERDE DOCUMENTEN

The research presented in this thesis was financially supported by European Research Council (ERC) advanced grant 293445, OnQview, a Roche Innovation Fund grant, a POINTING grant

We investigated anti-EGFR monoclonal antibodies imgatuzumab and cetuximab–induced internalization and membranous turnover of EGFR, and whether this affected

Preclinical positron emission tomography (PET) imaging revealed a mismatch between in vivo epidermal growth factor receptor (EGFR) expression and EGFR antibody tracer tumor

In addition, activation of HER3 is associated with resistance to anticancer agents such as PI3K-AKT pathway kinase inhibitors in human breast cancer cell lines (Chakrabarty et

HER3 expression in BT474 tumors remained unchanged after lapatinib therapy, as measured by IHC and Western blot (Fig. A) Membranous HER3 expression levels in BT474, SKBR3 and

A panel of EGFR wild-type lung adenocarcinoma cell lines, including a KRAS wild-type (H322), 3 KRAS mutant (A549, H358, and H460), and a KRAS wild-type adenocarcinoma-

Zandgronden met dik cultuurdek leemarm tot zwak lemig grof zand Zandgronden met matig dik cultuurdek leemarm en zwak lemig fijn zand Zandgronden met matig dik cultuurdek sterk

is a popular sensory marketing tool in the field of experiential marketing, applying such marketing techniques is a challenging and costly exercise for marketers,