Evaluation of integrin αvβ6 cystine knot PET tracers to detect cancer and idiopathic pulmonary fibrosis

Richard H Kimura, Ling Wang, Bin Shen, Li Huo, Willemieke Tummers, Fabian V Filipp, Haiwei Henry Guo, Thomas Haywood, Lotfi Abou-Elkacem, Lucia Baratto, Frezghi Habte, Rammohan Devulapally, Timothy H Witney, Yan Cheng, Suhas Tikole, Subhendu Chakraborti, Jay Nix, Christopher A Bonagura, Negin Hatami, Joshua J Mooney, Tushar Desai, Scott Turner, Richard S Gaster, Andrea Otte, Brendan C Visser, George A Poultsides, Jeffrey Norton, Walter Park, Mark Stolowitz, Kenneth Lau, Eric Yang, Arutselvan Natarajan, Ohad Ilovich, Shyam Srinivas, Ananth Srinivasan, Ramasamy Paulmurugan, Juergen Willmann, Frederick T Chin, Zhen Cheng, Andrei Iagaru, Fang Li, Sanjiv S Gambhir, Richard H Kimura, Ling Wang, Bin Shen, Li Huo, Willemieke Tummers, Fabian V Filipp, Haiwei Henry Guo, Thomas Haywood, Lotfi Abou-Elkacem, Lucia Baratto, Frezghi Habte, Rammohan Devulapally, Timothy H Witney, Yan Cheng, Suhas Tikole, Subhendu Chakraborti, Jay Nix, Christopher A Bonagura, Negin Hatami, Joshua J Mooney, Tushar Desai, Scott Turner, Richard S Gaster, Andrea Otte, Brendan C Visser, George A Poultsides, Jeffrey Norton, Walter Park, Mark Stolowitz, Kenneth Lau, Eric Yang, Arutselvan Natarajan, Ohad Ilovich, Shyam Srinivas, Ananth Srinivasan, Ramasamy Paulmurugan, Juergen Willmann, Frederick T Chin, Zhen Cheng, Andrei Iagaru, Fang Li, Sanjiv S Gambhir

Abstract

Advances in precision molecular imaging promise to transform our ability to detect, diagnose and treat disease. Here, we describe the engineering and validation of a new cystine knot peptide (knottin) that selectively recognizes human integrin αvβ6 with single-digit nanomolar affinity. We solve its 3D structure by NMR and x-ray crystallography and validate leads with 3 different radiolabels in pre-clinical models of cancer. We evaluate the lead tracer's safety, biodistribution and pharmacokinetics in healthy human volunteers, and show its ability to detect multiple cancers (pancreatic, cervical and lung) in patients at two study locations. Additionally, we demonstrate that the knottin PET tracers can also detect fibrotic lung disease in idiopathic pulmonary fibrosis patients. Our results indicate that these cystine knot PET tracers may have potential utility in multiple disease states that are associated with upregulation of integrin αvβ6.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Summary of the knottin PET ligand. a Primary structure of lead candidate R01-MG. The engineered active loop-1 is shown in blue. The integrin avβ6 core binding motif, RTDLxxL, is shown in black. The framework residues are shown in gray. Cysteine residues are shown in yellow and the pattern of disulfide bonds are indicated by the connecting lines. b Ensemble of 10 lowest energy three-dimensional 1H NMR of R01-MG structures. c Crystal structure of non-radioactive reference standard, [19F]FP-R01-MG-F2, at <1 Å. The carbonyl oxygen of the ester is shown in cyan. The fluorine atom is shown in red. The methyl carbon is shown in black. The insert on top shows the RP-HPLC trace of R01-MG, and the radio-RP-HPLC trace of the purified clinical-grade PET tracer [18F]FP-R01-MG-F2, respectively. The product is indicated by the asterisk. The insert on the bottom shows b the equilibrium binding curve of between R01-MG expressed on yeast surface and soluble human integrin αvβ6 (KD = 1.24 ± 0.21 nM S.D.), and c the competition binding between unlabeled R01-MG (IC50 = 0.61 ± 0.31 nM S.D., circles) or the N-terminus labeled version [19F]FP-R01-MG-F2 (IC50 = 0.56 ± 0.46 nM S.D., squares) vs. yeast surface expressed R01-MG, respectively. d Comparison at 1 h post injection of the top 3 [64Cu]DOTA-labeled PET tracer candidates, R01 (black), R01-MR (white) and R01-MG (gray). e Comparison at 1 h post injection of the [18F]FP labeled lead candidate R01-MG in integrin αvβ6 positive (cyan) vs. αvβ6 negative tumor models (magenta, dashed circle in the figure). In vivo validation of the [68Ga]NODAGA-R01-MG at 1 h post injection (yellow). f Volume rendered PET/CT images of [18F]FP-R01-MG (cyan) and [68Ga]NODAGA-R01-MG (yellow) in integrin avβ6 positive models at 1 h post injection. df The cyan arrows point to the tumor. The letters K and B represent the kidneys and bladder, respectively. (df and Table 3, below) Box colors are colored legends that cross-relate the images to the quantification table
Fig. 2
Fig. 2
[18F]FP-R01-MG-F2 PET imaging of five healthy human volunteers. a Representative whole-body [18F]FP-R01-MG-F2 maximum intensity projection (MIP) PET images of a healthy volunteer (50-year-old male) show the biodistribution of PET tracer at ~ 5, 60, and 120 min post injection (p.i.). Focal uptake near the elbow and between the legs are the site of intravenous injection, and a tube containing the reference calibration standard, respectively. b Axial and coronal PET/CT images of the same healthy volunteer at 1 h p.i. Close-ups of the chest and abdominal regions show the heart (SUVmean ~ 0.9), liver (SUVmean ~ 0.8), lung (SUVmean ~ 1.3), pancreas (SUVmean ~ 1.9), stomach (SUVmean ~ 12.6), small intestines (SUVmean ~ 7.8), kidneys (SUVmean ~ 14.3), and bladder (5.9). Accumulation of the tracer is evident in the pituitary gland (SUVmean ~ 5). c H&E staining (top left) and integrin αvβ6 (top right and bottom) immunohistochemical analysis of healthy stomach and small bowel tissue, where uptake was relatively high (Supplementary Table 7, below), shows expression of integrin αvβ6 on the luminal (Lu) side of these organs. Scale bars on the 1x and 10x images represent 2.5 cm and 250 mm, respectively
Fig. 3
Fig. 3
[18F]FP-R01-MG-F2 PET imaging of pancreatic cancer. a [18F]FP- R01-MG-F2 and (b) [18F]FDG images, from left to right, show MIP, CT, PET/CT (axial and coronal) and volume rendered PET/CT (full and magnified) images of a 71-year-old female pancreatic cancer patient at ~60 min post injection. The liver, kidneys, small intestines, stomach and spleen are denoted by Lv, Kd, SI, St, and Sp, respectively. The cyan arrow points to the tumor. a Accumulation of [18F]FP- R01-MG-F2 at the head of the tumor is shown by the cyan arrow (SUVmean = 6.3). PET/CT images demonstrate several regions of relatively high accumulation including the kidneys, the main clearance route, and the stomach (St), small intestines (SI) where integrin αvβ6 is expressed. b An area of focal [18F]FDG uptake (SUVmean = 2.3) which is located within the tumor coincides with a biliary stent (white tube) that is apparent in the CT image. The tracer is seen draining out of the kidney (Kd) through the ureter and collecting in the bladder in the MIP, and volume rendered PET/CT images. c From left to right, H&E staining and IHC analysis of the resected pancreatic mass, which included some healthy pancreatic tissue. A section of normal pancreas (left) and a section of malignant tissue (right) are shown. Scale bars on the 1x and 10x images represent 2.5 cm and 250 mm, respectively
Fig. 4
Fig. 4
[68Ga]NODAGA-R01-MG biodistribution and PET imaging of pancreatic cancer. a Sequential MIP PET images of a 69-year-old female study subject show the biodistribution of the tracer for up to ~1 h after intravenous administration. b PET/CT image of the brain shows focal uptake of the tracer in the pituitary gland. c ROI analysis of the representative patient shown above. The following organs are shown: brain (Br), pituitary gland (Pt), breast (Brs), lower large intestine (LLI), small intestine (SI), stomach (St), upper large intestine (ULI), heart wall (HW), heart contents (HC), kidney (Kd), liver (Lv), lung (Ln), muscle (Ms), red marrow (RM), osteogenic cells (Bn), skin (Sk), spleen (Sp), thymus (Tm), thyroid (Tr), bladder (Bl), and total body (TB). d Coronal and axial PET/CT images (left and center) and volume rendered PET/CT image (top right) acquired from a patient diagnosed with pancreatic cancer. The arrows indicate the location of the tumor. High uptake is observed throughout most of the pancreas including the head (SUVmean ~ 3.1), uncinate process, neck and tail (SUVmean ~ 4.4). Comparatively, the SUVmean = 1.8 ± 0.5 S.D. for normal pancreas (n = 5). e IHC confirms integrin αvβ6 expression in the viable part of the tumor. Red box represents area shown at 14x magnification in image below. Pathology report indicated a large amount of necrosis in the tumor. Scale bars on the unmagnified and 14x images represent 2.5cm and 250 mm, respectively. Source data for panel c are provided in a Source Data file
Fig. 5
Fig. 5
[68Ga]NODAGA-R01-MG PET imaging of cervical cancer and lung cancer. a Coronal (top left), sagittal (top right) and axial (bottom left) planes of PET/CT images of a 47-year-old cervical cancer patient shows tracer accumulation in the tumor (cyan arrow). Bl, Kd, Lv, SI, Sp, and St refer to the bladder, kidneys, liver, small intestine, spleen and stomach, respectively. b H&E staining and IHC analysis confirm expression of integrin αvβ6 by the larger diffuse cancer cells. Integrin αvβ6 was not expressed by the normal cells of the cervix surrounding the tumor. Red box represents area shown at 15x magnification in image below. Scale bars on the 1x and 15x images represent 2.5 cm and 250 mm, respectively. c Coronal (left), axial (middle) and sagittal (right) planes of PET/CT Images of a 70-year-old patient diagnosed with poorly differentiated squamous cell carcinoma of the lung (SUVmean = 2.2). The cyan arrow points to the tumor. The healthy lung and heart are indicated by Ln and H, respectively
Fig. 6
Fig. 6
[18F]FP-R01-MG-F2 PET imaging of lungs: IPF patients and healthy volunteers. a PET/CT images of IPF-5, a 72-year-old male IPF patient with a definite-UIP pattern, as indicated by peripheral, basilar predominant reticulation and honeycombing (CT, top, cyan arrows). Correspondingly, the PET tracer is concentrated in regions of greatest fibrosis in the areas of high reticulation and honeycombing in the lung bases (PET, middle and PET/CT, bottom, cyan arrows). In contrast, relatively healthy regions of the upper and anterior lungs show low tracer accumulation (PET/CT, bottom, white arrows). b PET/CT images of a 72-year-old male IPF patient with a single transplanted (2016) left lung (white arrows), which is relatively devoid of the PET tracer. The fibrotic right lung (IPF) shows elevated PET tracer levels in areas that also correspond to the highly fibrosed regions demonstrated on CT (PET/CT, bottom, cyan arrows). c PET/CT images of the lungs of five healthy volunteers, varying in age from 20- to 48-year old. The group demonstrated a range of lower [18F]FP- R01-MG-F2 accumulation in their lungs. d The dashed lines represent histograms of SUVs found within ROI contours of total lung. The upper histogram represents the healthy volunteers (HV-1 to HV-5). The lower histogram (dashed blue line) represents patient IPF-5 shown in panel A. For comparative purposes, the solid black curves in both (top and bottom graphs) represent baseline-20 SUVmean and normal distribution (BL-20 ND) derived from the two youngest healthy volunteers in their 20 s (HV-3 and HV-5)
Fig. 7
Fig. 7
Mean SUVs of healthy volunteers and IPF patients. Mean SUVs from healthy volunteers and IPF patients are shown as circles and triangles, respectively. The single transplanted lung in patient IPF-4 is shown as a circle (far right). The right/left mean SUVs for patient IPF-3 were identical and appear as a single triangle (Supplemental Table 13). The right/left correlation is 0.90 and 0.70 for HVs and IPF patients, respectively. The average SUVmeans ± S.D.s for the right lungs of HVs (n = 5) and the IPF group (n = 6) is 0.92 ± 0.33 and 2.04 ± 0.91, respectively (p = 0.0087). Source data are referred to in the Source Data File

References

    1. Raghu G, et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am. J. Respir. Crit. care Med. 2011;183:788–824. doi: 10.1164/rccm.2009-040GL.
    1. Saini G, et al. alphavbeta6 integrin may be a potential prognostic biomarker in interstitial lung disease. Eur. Respir. J. 2015;46:486–494. doi: 10.1183/09031936.00210414.
    1. Ruoslahti E. RGD and other recognition sequences for integrins. Annu Rev. Cell Dev. Biol. 1996;12:297–715. doi: 10.1146/annurev.cellbio.12.1.697.
    1. Kimura RH, Cheng Z, Gambhir SS, Cochran JR. Engineered knottin peptides: a new class of agents for imaging integrin expression in living subjects. Cancer Res. 2009;69:2435–2442. doi: 10.1158/0008-5472.CAN-08-2495.
    1. Haubner R, Maschauer S, Prante O. PET radiopharmaceuticals for imaging integrin expression: tracers in clinical studies and recent developments. Biomed. Res Int. 2014;2014:1–17.
    1. Gaertner FC, Kessler H, Wester HJ, Schwaiger M, Beer AJ. Radiolabelled RGD peptides for imaging and therapy. Eur. J. Nucl. Med. Mol. imaging. 2012;39:126–138. doi: 10.1007/s00259-011-2028-1.
    1. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. 2002;110:673–687. doi: 10.1016/S0092-8674(02)00971-6.
    1. Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat. Rev. Cancer. 2010;10:9–22. doi: 10.1038/nrc2748.
    1. Reed NI, et al. The alphavbeta1 integrin plays a critical in vivo role in tissue fibrosis. Sci. Transl. Med. 2015;7:288ra279. doi: 10.1126/scitranslmed.aaa5094.
    1. Kingwell K. Fibrotic diseases: New tools to target an elusive integrin. Nat. Rev. Drug Discov. 2015;14:460. doi: 10.1038/nrd4672.
    1. Henderson NC, et al. Targeting of alphav integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat. Med. 2013;19:1617–1624. doi: 10.1038/nm.3282.
    1. Chen H, Niu G, Wu H, Chen X. Clinical application of radiolabeled RGD peptides for PET imaging of integrin alphavbeta3. Theranostics. 2016;6:78–92. doi: 10.7150/thno.13242.
    1. John AE, et al. Preclinical SPECT/CT imaging of alphavbeta6 integrins for molecular stratification of idiopathic pulmonary fibrosis. J. Nucl. Med.: Off. Publ., Soc. Nucl. Med. 2013;54:2146–2152. doi: 10.2967/jnumed.113.120592.
    1. Bandyopadhyay A, Raghavan S. Defining the role of integrin alphavbeta6 in cancer. Curr. Drug Targets. 2009;10:645–652. doi: 10.2174/138945009788680374.
    1. Breuss JM, et al. Expression of the beta 6 integrin subunit in development, neoplasia and tissue repair suggests a role in epithelial remodeling. J. Cell Sci. 1995;108:2241–2251.
    1. Sipos B, et al. Immunohistochemical screening for beta6-integrin subunit expression in adenocarcinomas using a novel monoclonal antibody reveals strong up-regulation in pancreatic ductal adenocarcinomas in vivo and in vitro. Histopathology. 2004;45:226–236. doi: 10.1111/j.1365-2559.2004.01919.x.
    1. Maubant S, et al. Expression of alpha V-associated integrin beta subunits in epithelial ovarian cancer and its relation to prognosis in patients treated with platinum-based regimens. J. Mol. Histol. 2005;36:119–129. doi: 10.1007/s10735-004-4273-0.
    1. Orimoto AM, et al. High numbers of human skin cancers express MMP2 and several integrin genes. J. Cutan. Pathol. 2008;35:285–291. doi: 10.1111/j.1600-0560.2007.00810.x.
    1. Bates RC, et al. Transcriptional activation of integrin beta6 during the epithelial-mesenchymal transition defines a novel prognostic indicator of aggressive colon carcinoma. J. Clin. Invest. 2005;115:339–347. doi: 10.1172/JCI200523183.
    1. Elayadi AN, et al. A peptide selected by biopanning identifies the integrin alphavbeta6 as a prognostic biomarker for nonsmall cell lung cancer. Cancer Res. 2007;67:5889–5895. doi: 10.1158/0008-5472.CAN-07-0245.
    1. Hazelbag S, et al. Overexpression of the alpha v beta 6 integrin in cervical squamous cell carcinoma is a prognostic factor for decreased survival. J. Pathol. 2007;212:316–324. doi: 10.1002/path.2168.
    1. Zhang ZY, et al. Integrin alphanvbeta6 acts as a prognostic indicator in gastric carcinoma. Clin. Oncol. (R. Coll. Radio.). 2008;20:61–66. doi: 10.1016/j.clon.2007.09.008.
    1. Yang GY, et al. Integrin alpha v beta 6 mediates the potential for colon cancer cells to colonize in and metastasize to the liver. Cancer Sci. 2008;99:879–887. doi: 10.1111/j.1349-7006.2008.00762.x.
    1. Tummers, W. S. et al. Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma. Oncotarget8, 56816–56828 (2017).
    1. Hausner SH, DiCara D, Marik J, Marshall JF, Sutcliffe JL. Use of a peptide derived from foot-and-mouth disease virus for the noninvasive imaging of human cancer: generation and evaluation of 4-[18F]fluorobenzoyl A20FMDV2 for in vivo imaging of integrin alphavbeta6 expression with positron emission tomography. Cancer Res. 2007;67:7833–7840. doi: 10.1158/0008-5472.CAN-07-1026.
    1. Kimura RH, et al. Pharmacokinetically stabilized cystine knot peptides that bind alpha-v-beta-6 integrin with single-digit nanomolar affinities for detection of pancreatic cancer. Clin. Cancer Res. 2012;18:839–849. doi: 10.1158/1078-0432.CCR-11-1116.
    1. Altmann A, et al. Identification of a Novel ITGalphavbeta6-Binding Peptide Using Protein Separation and Phage Display. Clin. Cancer Res.: Off. J. Am. Assoc. Cancer Res. 2017;23:4170–4180. doi: 10.1158/1078-0432.CCR-16-3217.
    1. Horan GS, et al. Partial inhibition of integrin alpha(v)beta6 prevents pulmonary fibrosis without exacerbating inflammation. Am. J. Respir. Crit. care Med. 2008;177:56–65. doi: 10.1164/rccm.200706-805OC.
    1. Munger JS, et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell. 1999;96:319–328. doi: 10.1016/S0092-8674(00)80545-0.
    1. Ignotz RA, Massague J. Transforming growth factor-beta stimulates the expression of fibronectin and collagen and their incorporation into the extracellular matrix. J. Biol. Chem. 1986;261:4337–4345.
    1. Kimura RH, Levin AM, Cochran FV, Cochran JR. Engineered cystine knot peptides that bind alphavbeta3, alphavbeta5, and alpha5beta1 integrins with low-nanomolar affinity. Proteins. 2009;77:359–369. doi: 10.1002/prot.22441.
    1. Hackel BJ, Kimura RH, Gambhir SS. Use of (64)Cu-labeled fibronectin domain with EGFR-overexpressing tumor xenograft: molecular imaging. Radiology. 2012;263:179–188. doi: 10.1148/radiol.12111504.
    1. Colgrave ML, Craik DJ. Thermal, chemical, and enzymatic stability of the cyclotide kalata B1: the importance of the cyclic cystine knot. Biochemistry. 2004;43:5965–5975. doi: 10.1021/bi049711q.
    1. Shrout PE, Fleiss JL. Intraclass correlations: uses in assessing rater reliability. Psychol. Bull. 1979;86:420–428. doi: 10.1037/0033-2909.86.2.420.
    1. Reed CE, Barbee RA. Pigeon-breeders’ lung: a newly observed interstitial pulmonary disease. Jama. 1965;193:261–265. doi: 10.1001/jama.1965.03090040005001.
    1. Ley B, Collard HR. Epidemiology of idiopathic pulmonary fibrosis. Clin. Epidemiol. 2013;5:483–492. doi: 10.2147/CLEP.S54815.
    1. Xiong JP, et al. Crystal structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand. Sci. (New Y., N. Y.) 2002;296:151–155. doi: 10.1126/science.1069040.
    1. Hausner SH, Bauer N, Hu LY, Knight LM, Sutcliffe JL. The effect of bi-terminal PEGylation of an integrin alphavbeta(6)-targeted (1)(8)F peptide on pharmacokinetics and tumor uptake. J. Nucl. Med.: Off. Publ., Soc. Nucl. Med. 2015;56:784–790. doi: 10.2967/jnumed.114.150680.
    1. Kraft S, et al. Definition of an unexpected ligand recognition motif for alphav beta6 integrin. J. Biol. Chem. 1999;274:1979–1985. doi: 10.1074/jbc.274.4.1979.
    1. Nothelfer EM, et al. Identification and characterization of a peptide with affinity to head and neck cancer. J. Nucl. Med.: Off. Publ., Soc. Nucl. Med. 2009;50:426–434. doi: 10.2967/jnumed.108.058123.
    1. Willmann JK, et al. Targeted contrast-enhanced ultrasound imaging of tumor angiogenesis with contrast microbubbles conjugated to integrin-binding knottin peptides. J. Nucl. Med.: Off. Publ., Soc. Nucl. Med. 2010;51:433–440. doi: 10.2967/jnumed.109.068007.
    1. Zhang C, et al. A cystine knot peptide targeting integrin alphavbeta6 for photoacoustic and fluorescence imaging of tumors in living subjects. J. Nucl. Med.: Off. Publ., Soc. Nucl. Med. 2016;57:1629–1634. doi: 10.2967/jnumed.115.169383.
    1. Chin FT, et al. First experience with clinical-grade ([18F]FPP(RGD(2)): an automated multi-step radiosynthesis for clinical PET studies. Mol. Imaging Biol.: MIB: Off. Publ. Acad. Mol. Imaging. 2012;14:88–95. doi: 10.1007/s11307-011-0477-3.
    1. Breuss JM, Gillett N, Lu L, Sheppard D, Pytela R. Restricted distribution of integrin beta 6 mRNA in primate epithelial tissues. The journal of histochemistry and cytochemistry: official journal of the Histochemistry. Society. 1993;41:1521–1527.
    1. Sheppard D. Roles of alphav integrins in vascular biology and pulmonary pathology. Curr. Opin. cell Biol. 2004;16:552–557. doi: 10.1016/j.ceb.2004.06.017.
    1. Henderson NC, Sheppard D. Integrin-mediated regulation of TGFbeta in fibrosis. Biochim. et. Biophys. acta. 2013;1832:891–896. doi: 10.1016/j.bbadis.2012.10.005.
    1. Popov Y, et al. Integrin alphavbeta6 is a marker of the progression of biliary and portal liver fibrosis and a novel target for antifibrotic therapies. J. Hepatol. 2008;48:453–464. doi: 10.1016/j.jhep.2007.11.021.
    1. Conroy KP, Kitto LJ, Henderson NC. alphav integrins: key regulators of tissue fibrosis. Cell tissue Res. 2016;365:511–519. doi: 10.1007/s00441-016-2407-9.
    1. Raghu, G. et al. American Thoracic Society: ATS Conference 2018 (San Diego CA USA, 2018).
    1. Slack, R. J. et al. The International Colloquim on Lung and Airway Fibrosis (ICLAF). (Dublin, Ireland, 2016).
    1. Rosas IO, et al. Interstitial lung disease: NHLBI Workshop on the Primary Prevention of Chronic Lung Diseases. Ann. Am. Thorac. Soc. 2014;11(Suppl 3):S169–S177. doi: 10.1513/AnnalsATS.201312-429LD.
    1. Doyle TJ, Hunninghake GM, Rosas IO. Subclinical interstitial lung disease: why you should care. Am. J. Respir. Crit. care Med. 2012;185:1147–1153. doi: 10.1164/rccm.201108-1420PP.
    1. Margadant C, Sonnenberg A. Integrin-TGF-beta crosstalk in fibrosis, cancer and wound healing. EMBO Rep. 2010;11:97–105. doi: 10.1038/embor.2009.276.
    1. Farnoud MR, Veirana N, Derome P, Peillon F, Li JY. Adenomatous transformation of the human anterior pituitary is associated with alterations in integrin expression. Int. J. cancer. 1996;67:45–53. doi: 10.1002/(SICI)1097-0215(19960703)67:1<45::AID-IJC10>;2-B.
    1. Recouvreux MV, Camilletti MA, Rifkin DB, Diaz-Torga G. The pituitary TGFbeta1 system as a novel target for the treatment of resistant prolactinomas. J. Endocrinol. 2016;228:R73–R83. doi: 10.1530/JOE-15-0451.
    1. Farrell WE. Pituitary tumours: findings from whole genome analyses. Endocr.-Relat. cancer. 2006;13:707–716. doi: 10.1677/erc.1.01131.
    1. Paulus W, Baur I, Schuppan D, Roggendorf W. Characterization of integrin receptors in normal and neoplastic human brain. Am. J. Pathol. 1993;143:154–163.
    1. Ying SY, et al. Type beta transforming growth factor (TGF-beta) is a potent stimulator of the basal secretion of follicle stimulating hormone (FSH) in a pituitary monolayer system. Biochem. Biophys. Res. Commun. 1986;135:950–956. doi: 10.1016/0006-291X(86)91020-X.
    1. Fujiwara K, Ikeda H, Yoshimoto T. Immunohistochemical demonstration of TGF-beta-receptor type II in human pituitary adenomas. Acta Histochem. 1995;97:445–454. doi: 10.1016/S0065-1281(11)80071-1.
    1. Munger JS, Sheppard D. Cross talk among TGF-beta signaling pathways, integrins, and the extracellular matrix. Cold Spring Harb. Perspect. Biol. 2011;3:a005017. doi: 10.1101/cshperspect.a005017.
    1. Stabin MG, Sparks Rb Fau - Crowe E, Crowe E. OLINDA/EXM: the second-generation personal computer software for internal dose assessment in nuclear medicine. J. Nucl. Med. 2005;46:1023–1027.
    1. Kimura RH, Cheng Z, Gambhir SS, Cochran JR. Engineered knottin peptides: a new class of agents for imaging integrin expression in living subjects. Cancer Res. 2009;69:2435–2442. doi: 10.1158/0008-5472.CAN-08-2495.
    1. Filipp FabianV, Sattler Michael. Conformational Plasticity of the Lipid Transfer Protein SCP2. Biochemistry. 2007;46:7980–7991. doi: 10.1021/bi6025616.
    1. Filipp FabianV, Scott DavidA, Ronai Ze’evA, Osterman AndreiL, Smith JeffreyW. Reverse TCA cycle flux through isocitrate dehydrogenases 1 and 2 is required for lipogenesis in hypoxic melanoma cells. Pigment Cell & Melanoma Research. 2012;25:375–383. doi: 10.1111/j.1755-148X.2012.00989.x.
    1. Alice Douangamath FabianV, et al. Topography for Independent Binding of α-Helical and PPII-Helical Ligands to a Peroxisomal SH3 Domain. Molecular Cell. 2002;10:1007–1017. doi: 10.1016/S1097-2765(02)00749-9.
    1. Stanley WillA, et al. Recognition of a Functional Peroxisome Type 1 Target by the Dynamic Import Receptor Pex5p. Molecular Cell. 2006;24:653–663. doi: 10.1016/j.molcel.2006.10.024.
    1. Shannon L, et al. Probing lipid- and drug-binding domains with fluorescent dyes. Bioorganic & Medicinal Chemistry. 2008;16:1162–1173. doi: 10.1016/j.bmc.2007.10.080.
    1. Delaglio, F. et al. NMRPipe: A multidimensional spectral processing system based on UNIX pipes. Journal of Biomolecular NMR, (1995) 6
    1. Johnson BruceA, Blevins RichardA. NMR View: A computer program for the visualization and analysis of NMR data. Journal of Biomolecular NMR. 1994;4:603–614. doi: 10.1007/BF00404272.
    1. Peng C, Unger SW, Filipp FV, Sattler M, Szalma S. Automated evaluation of chemical shift perturbation spectra: New approaches to quantitative analysis of receptor-ligand interaction NMR spectra. J Biomol NMR. 2004;29:491–504. doi: 10.1023/B:JNMR.0000034351.37982.9e.
    1. Filipp FabianV, Sinha Neeraj, Jairam Lena, Bradley Joel, Opella StanleyJ. Labeling strategies for 13C-detected aligned-sample solid-state NMR of proteins. Journal of Magnetic Resonance. 2009;201:121–130. doi: 10.1016/j.jmr.2009.08.012.
    1. Scott DavidA, et al. Comparative Metabolic Flux Profiling of Melanoma Cell Lines. Journal of Biological Chemistry. 2011;286:42626–42634. doi: 10.1074/jbc.M111.282046.
    1. Günert P. Automated NMR structure calculation with CYANA. Methods Mol Biol. 2004;278:353–378. doi: 10.1385/1-59259-809-9:353.
    1. Thompson AC, Westbrook EM, Lavender WM, Nix JC. A large area CMOS detector for shutterless collection of x-ray diffraction data. J. Phys.: Conf. Ser. 2014;493:012019.
    1. Kabsch W. XDS. Acta Crystallogr. Sect. D. 2010;D66:125–132. doi: 10.1107/S0907444909047337.
    1. Collaborative_Computational_Project. The CCP4 suite: programs for protein crystallography. Acta Crystallogr. D. Biol. Crystallogr. 1994;50:760–763. doi: 10.1107/S0907444994003112.
    1. Adams Paul D., Grosse-Kunstleve Ralf W., Hung Li-Wei, Ioerger Thomas R., McCoy Airlie J., Moriarty Nigel W., Read Randy J., Sacchettini James C., Sauter Nicholas K., Terwilliger Thomas C. PHENIX: building new software for automated crystallographic structure determination. Acta Crystallographica Section D Biological Crystallography. 2002;58(11):1948–1954. doi: 10.1107/S0907444902016657.
    1. McCoy Aj, et al. Phaser crystallographic software. J. Appl. Crystallogr. 2007;40:658–674. doi: 10.1107/S0021889807021206.
    1. Perrakis A., Harkiolaki M., Wilson K. S., Lamzin V. S. ARP/wARP and molecular replacement. Acta Crystallogr. D 57, 1445-1450 (2001).
    1. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D 60, 2126–2132 (2004).

Source: PubMed

3
Sottoscrivi