Tumor Associated Inflammation: Biology & Imaging
Heike Elisabeth Daldrup-Link1

1Department of Radiology, Stanford University, Stanford, CA, United States

Synopsis

Many malignant tumors, including breast cancer, lung cancer, colon cancer, pancreatic cancer, lymphomas, sarcomas, neuroblastomas and (among many others) are associated with a pro-inflammatory tumor microenvironment, characterized by infiltration of leukocytes whereas increases in some leukocyte subsets parallels disease progression and worse clinical outcomes (1-4). Tumor-associated macrophages (TAMs) play a key role in this context: M2-TAM phenotypes promote tumor growth and enhance pulmonary metastasis by high-level expression of epidermal growth factor (EGF) and activation of EGF-regulated signaling pathways critical for invasive tumor growth and metastatic dissemination. Conversely, M1-TAM phenotypes mediate tumor cell phagocytosis and tumor growth inhibition directly or indirectly. New therapeutic drugs that target TAM are currently being developed and are starting to enter the clinic. Thus, it becomes increasingly important to identify patients whose tumors are heavily infiltrated by TAM, in order to stratify these patients to TAM-directed immunotherapies and monitor treatment response (4-6). Unfortunately, existing clinical imaging technologies do not provide a good method for evaluating response to immunotherapies because most immune modulating therapeutics do not cause changes in tumor size, at least not in the immediate post-treatment time period, but rather changes in the immune cell composition of the tumor (7-9). Therefore, as novel immunotherapies are being integrated with classical chemotherapy (7-9), it becomes increasingly important to monitor the immune cell signature of malignant tumors. To serve this goal, an imaging test is advantageous over invasive biopsy because it is non-invasive, covers the whole tumor and can repeatedly interrogate treatment effects on the complex cross talk between innate and adaptive immune responses in vivo, in patients.

Historically, molecular imaging tests have focused on imaging cancer cells, tumor microvascular characteristics and the extracellular matrix surrounding cancer (10-12). The tumor microenvironment has not been a major focus for the development of novel imaging technologies. In the past, our team has pioneered a number of novel imaging approaches for cancer MR imaging (13-19), PET imaging (20-26), and immune cell imaging (13,14,23,27,28), including translational MR imaging tests for in vivo detection of TAM with the FDA-approved iron supplement ferumoxytol (Feraheme) (29,30). Ferumoxytol is a colloid-based USPIO with a hydrodynamic diameter of 28–32 nm, consisting of an iron oxide core with a size of 6.4 –7.2 nm and a carboxymethyldextran coat. At 20 MHz and 39°C, ferumoxytol has an r1 relaxivity of 38 mM–1s–1 and an r2 relaxivity of 83 mM–1s–1 and can therefore be used as a contrast agent for MRI. In fact, ferumoxytol is the only FDA-approved iron oxide nanoparticle compound to date, which can be offered to patients for non-invasive TAM imaging through “off label” use as a contrast agent. Unlike larger SPIO (> 50 nm), intravenously injected ferumoxytol nanoparticles (< 50 nm) transiently escape RES-phagocytosis in liver, spleen and bone marrow, which leads to a prolonged blood half-life. Ferumoxytol nanoparticles then slowly “leak” across hyperpermeable tumor microvessels into the tumor interstitium, where they are slowly phagocytosed by TAM, a process that takes several hours (29). At 24 hours p.i., the nanoparticles are compartmentalized intracellulary in TAM (Fig. 1). Following intravenous administration, ferumoxytol nanoparticles show an initial perfusion effect of tumor tissue, followed by transendothelial leak and phagocytosis by TAM via the “enhanced permeability and retention (EPR) effect”, which results in a marked hypointense signal effect on delayed T1- and T2-weighted MR images (29). This signal effect can be used to noninvasively track the degree of macrophage infiltration in a tumor before and after TAM-modulating immunotherapies. Our clinical MR imaging studies after intravenous injection of iron oxide nanoparticles demonstrate unique enhancement properties of glioblastomas, lymphomas and sarcomas in the presence of TAM-mediated inflammation, providing a novel, immediately clinically applicable imaging test to noninvasively track innate immune responses from macrophages in patients (30-32). Intracellular nanoparticles in TAMs exert a T2-signal effect on delayed MR scans, several hours to days after nanoparticle administration, with minimal or no hyperintense T1 effect due to lack of interactions with protons. This “decoupling” of T1- and T2-signal effects is different compared to the additive T1- and T2-signal effects of free iron oxides in vessels or tumor necrosis and can be used as a non-invasive imaging indicator for iron-labeled TAMs (33). Recent developments in macrophage-targeted PET-tracers may improve the specificity of iron oxide nanoparticle-based MR imaging through integrated PET/MR approaches. These novel TAM imaging concepts could represent a significant breakthrough for clinicians as a new means for risk stratification and as a new gold-standard imaging test for tracking treatment response in future TAM-directed immunotherapy trials, including planned first-in-man anti-CD47 immunotherapy trials in 2016.

Summary: in vivo imaging of distinct immune cell populations in malignant tumors should permeate our understanding of immune cell responses to cancer therapies, enable us to overcome the bottleneck of monitoring antitumor immune responses in vivo, improve patient stratification to tailored immune modulating therapies and aid in assigning non-responders to alternative treatment options. For example, patients with cancers with marked TAM infiltration could be directed to novel immune reprogramming therapies, while patients with node-negative tumors without significant leukocyte infiltration could be spared. TAM imaging approaches could also facilitate the development, monitoring and regulatory approval of new classes of immune-based drugs. Since clinical trials of new therapeutic drugs and combination therapies are expensive and take years to complete, novel TAM imaging approaches provide significant immediate value and impact. The ability to non-invasively and repetitively image several distinct immune cell populations in the tumor microenvironment will open opportunities for new discoveries in the area of cancer immunology and immunotherapy, and ultimately, help to improve long term treatment outcomes.

Figure Legend

Figure 1: In vivo biodistribution and MR signal effects of intravenously administered superparamagnetic iron oxide nanoparticles (SPIO) with hydrodynamic diameters in the order of 20-100 nm: After intravenous injection, SPIO initially distribute in the blood pool, where they exert a strong T1- and T2-effect. Due to their large size, nanoparticles do not extravasate across intact vascular endothelia in most normal organs. However, the nanoparticles leak across hyperpermeable sinus in organs of the reticuloendothelial system (RES), such as bone marrow, liver, spleen and lymph nodes, which leads to a hypointense signal effect on T2- and T2*-weighted MR images. Nanoparticle extravasation occurs at a relatively slower rate into the interstitium of malignant tumors, where SPIO are phagocytosed by tumor-associated macrophages (TAM). Therefore, tumors in RES organs can be depicted as T2-hyperintense lesions on relatively early postcontrast scans, but do show T2-enhancement on delayed postcontrast scans. Intracellular SPIO in TAM exert a hypo-intense MR signal effect on both T1- and T2-weighted MR images while extracellular SPIO in tumor necrosis demonstrate T1-hyperintense and T2-hypointense signal. Within macrophages, SPIO undergo a slow metabolization. Baseline MR signal intensities are regained after several weeks

Acknowledgements

This work was supported by grants from the National Institute of Health (NIH), the Eunice Kennedy Shriver National Institute of Child Health and Human Development (R01 HD081123A), the National Cancer Institute (R21CA176519 and R21CA190196) and the National Institute of Arthritis and Musculoskeletal and Skin Diseases (2R01AR054458). Parts of the described work here are covered under patent US6009342-A, licensed to the University of California in San Francisco, patent 13/923,962, licensed to Stanford University; and patent UK 13-005, licensed to the University of Bradford, UK and Stanford University. This work was also supported by colleagues and equipment resources at the Small Animal Imaging Facility and the Lucas Center at Stanford.

References

1. DeNardo, D.G. & Coussens, L.M. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res 9, 212 (2007).

2. DeNardo, D.G., et al. Leukocyte complexity in breast cancer predicts overall survival and functionally regulates response to chemotherapy. Cancer discovery 1, 54-67 (2011).

3. Ruffell, B., et al. Leukocyte composition of human breast cancer. Proc Natl Acad Sci U S A 109, 2796-2801 (2012).

4. Coussens, L.M., Zitvogel, L. & Palucka, A.K. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science 339, 286-291 (2013).

5. Anand, S. & Coussens, L.M. Manipulating microRNAs to regulate macrophage polarization in gliomas. Journal of the National Cancer Institute 106(2014).

6. Strachan, D.C., et al. CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8 T cells. Oncoimmunology 2, e26968 (2013).

7. Swartz, M.A., et al. Tumor microenvironment complexity: emerging roles in cancer therapy. Cancer Res 72, 2473-2480 (2012).

8. Shiao, S.L., Ganesan, A.P., Rugo, H.S. & Coussens, L.M. Immune microenvironments in solid tumors: new targets for therapy. Genes & development 25, 2559-2572 (2011).

9. Stewart, T.J. & Smyth, M.J. Improving cancer immunotherapy by targeting tumor-induced immune suppression. Cancer metastasis reviews 30, 125-140 (2011).

10. Smith, J.J., Sorensen, A.G. & Thrall, J.H. Biomarkers in imaging: realizing radiology's future. Radiology 227, 633-638 (2003).

11. Cai, W. & Chen, X. Multimodality molecular imaging of tumor angiogenesis. J Nucl Med 49 Suppl 2, 113S-128S (2008).

12. Oyen, W.J. & van der Graaf, W.T. Molecular imaging of solid tumors: exploiting the potential. Nat Rev Clin Oncol 6, 609-611 (2009).

13. Daldrup-Link, H. & Coussens, L.M. MR imaging of tumor-associated macrophages. Oncoimmunology 1, 507-509 (2012).

14. Daldrup-Link, H.E., et al. MR Imaging of Tumor Associated Macrophages with Clinically-Applicable Iron Oxide Nanoparticles. Clin Cancer Res (2011).

15. Daldrup-Link, H.E., et al. Quantification of breast tumor microvascular permeability with feruglose-enhanced MR imaging: initial phase II multicenter trial. Radiology 229, 885-892 (2003).

16. Daldrup-Link, H.E., Simon, G.H. & Brasch, R.C. Imaging of tumor angiogenesis: current approaches and future prospects. Current pharmaceutical design 12, 2661-2672 (2006).

17. Meier, R., et al. Breast cancers: MR imaging of folate-receptor expression with the folate-specific nanoparticle P1133. Radiology 255, 527-535 (2010).

18. Shi, Q., et al. Evaluation of the novel USPIO GEH121333 for MR imaging of cancer immune responses. Contrast Media & Molecular Imaging (in press).

19. Wang, Z.J., et al. MR imaging of ovarian tumors using folate-receptor-targeted contrast agents. Pediatr Radiol 38, 529-537 (2008).

20. Baek, S., et al. Exploratory clinical trial of (4S)-4-(3-[18F]fluoropropyl)-L-glutamate for imaging xC- transporter using positron emission tomography in patients with non-small cell lung or breast cancer. Clin Cancer Res 18, 5427-5437 (2012).

21. Hackel, B.J., Kimura, R.H. & Gambhir, S.S. Use of (64)Cu-labeled fibronectin domain with EGFR-overexpressing tumor xenograft: molecular imaging. Radiology 263, 179-188 (2012).

22. Jadvar, H., Alavi, A. & Gambhir, S.S. 18F-FDG uptake in lung, breast, and colon cancers: molecular biology correlates and disease characterization. J Nucl Med 50, 1820-1827 (2009).

23. James, M.L. & Gambhir, S.S. A molecular imaging primer: modalities, imaging agents, and applications. Physiological reviews 92, 897-965 (2012).

24. Lin, F.I., et al. Prospective comparison of combined (1)(8)F-FDG and (1)(8)F-NaF PET/CT vs. (1)(8)F-FDG PET/CT imaging for detection of malignancy. Eur J Nucl Med Mol Imaging 39, 262-270 (2012).

25. Natarajan, A., et al. Positron emission tomography of 64Cu-DOTA-Rituximab in a transgenic mouse model expressing human CD20 for clinical translation to image NHL. Molecular imaging and biology : MIB : the official publication of the Academy of Molecular Imaging 14, 608-616 (2012).

26. Nielsen, C.H., et al. PET imaging of tumor neovascularization in a transgenic mouse model with a novel 64Cu-DOTA-knottin peptide. Cancer Res 70, 9022-9030 (2010).

27. Hildebrandt, I.J. & Gambhir, S.S. Molecular imaging applications for immunology. Clin Immunol 111, 210-224 (2004).

28. Jha, P., et al. Monitoring of natural killer cell immunotherapy using noninvasive imaging modalities. Cancer Res 70, 6109-6113 (2010).

29. Daldrup-Link, H.E., et al. MRI of tumor-associated macrophages with clinically applicable iron oxide nanoparticles. Clin Cancer Res 17, 5695-5704 (2011).

30. Klenk, C., et al. Ionising radiation-free whole-body MRI versus (18)F-fluorodeoxyglucose PET/CT scans for children and young adults with cancer: a prospective, non-randomised, single-centre study. The lancet oncology 15, 275-285 (2014).

31. Iv, M., et al. Clinical applications of iron oxide nanoparticles for magnetic resonance imaging of brain tumors. Nanomedicine (Lond) 10, 993-1018 (2015).

32. Shi, Q., et al. Evaluation of the novel USPIO GEH121333 for MR imaging of cancer immune responses. Contrast Media Mol Imaging 8, 281-288 (2013).

33. Aghighi, M., et al. Imaging Tumor Necrosis with Ferumoxytol. PLoS One 10, e0142665 (2015).

Figures

See figure legend below text.



Proc. Intl. Soc. Mag. Reson. Med. 24 (2016)
0000