Abstract
In tribute to our friend and colleague Michael Robinson, we review his involvement in the identification, characterization and localization of the metallopeptidase glutamate carboxypeptidase II (GCPII), originally called NAALADase. While Mike was characterizing NAALADase in the brain, the protein was independently identified by other laboratories in human prostate where it was termed prostate specific membrane antigen (PSMA) and in the intestines where it was named Folate Hydrolase 1 (FOLH1). It was almost a decade to establish that NAALADase, PSMA, and FOLH1 are encoded by the same gene. The enzyme has emerged as a therapeutic target outside of the brain, with the most notable progress made in the treatment of prostate cancer and inflammatory bowel disease (IBD). PSMA-PET imaging with high affinity ligands is proving useful for the clinical diagnosis and staging of prostate cancer. A molecular radiotherapy based on similar ligands is in trials for metastatic castration-resistant prostate cancer (mCRPC). New PSMA inhibitor prodrugs that preferentially block kidney and salivary gland versus prostate tumor enzyme may improve the clinical safety of this radiotherapy. The wide clinical use of PSMA-PET imaging in prostate cancer has coincidentally led to clinical documentation of GCPII upregulation in a wide variety of tumors and inflammatory diseases, likely associated with angiogenesis. In IBD, expression of the FOLH1 gene that codes for GCPII is strongly upregulated, as is the enzymatic activity in diseased patient biopsies. In animal models of IBD, GCPII inhibitors show substantial efficacy, suggesting potential theranostic use of GCPII ligands for IBD.
Keywords: GCPII, PSMA, Folate Hydrolase, Prostate Cancer, Inflammatory Bowel Disease, Theranostics
Discovery and characterization of GCPII in brain, prostate and gut
Michael Robinson played a pivotal role in identifying, characterizing, and localizing the protein known to biochemists as glutamate carboxypeptidase II (GCPII), known to neuroscientists as NAALADase, known to the cancer world as prostate specific membrane antigen (PSMA), and known to intestinal researchers as folate hydrolase 1 (FOLH1). In this review, we will first detail Mike’s contribution in the early days of GCPII research, then describe the parallel work that identified the same protein in the prostate and gut. Finally, we will provide an overview of where that early work has led: therapeutic development of GCPII inhibitors and ligands outside of the brain for cancer and intestinal inflammation. It’s ironic that decades of work to develop small molecule inhibitors of GCPII to treat CNS disorders of presumed hyper-glutamatergic pathogenesis has actually led to their therapeutic promise outside of the nervous system, hence our title Looking for Drugs in All the Wrong Places.
As postdoctoral fellows in Joseph Coyle’s laboratory at Johns Hopkins, Mike and Randy Blakely characterized the enzymatic activity responsible for the cleavage of N-acetylaspartylglutamate (NAAG) to N-acetylaspartate (NAA) and glutamate [1]. At the time, NAAG had been proposed as an excitatory neurotransmitter as it was produced in the brain and mediated an excitatory effect [2,3], but Mike and Randy’s seminal 1986 publication [1] was the first to provide direct evidence of brain NAAG peptidase activity and liberation of glutamate from NAAG .
Through subsequent publications, the Blakely-Robinson duo pharmacologically characterized this membrane-bound metallopeptidase, which they named NAALADase [4,5]. These first characterizations of brain NAALADase described four-fold higher levels of activity in kidney than in in forebrain, with lower levels in other tissues tested including heart, lung, small intestine and adrenal gland [5]. Because the Coyle lab was focused on understanding the role of NAAG in synaptic transmission, the peripheral distribution of enzymatic activity was noted but not further studied by Mike or others in the lab.
Following his initial work characterizing NAAG hydrolysis, Mike played a key role in the isolation of the NAALADase protein, through his guidance and mentorship of Barbara Slusher, then a pharmacology graduate student in the Coyle lab. Mike and Barbara subsequently generated polyclonal antibodies and confirmed the localization of the protein in the brain and renal proximal tubules [6,7]. Later localization studies were extended to the cellular level and demonstrated expression by non-myelinating Schwann cells in peripheral nerves [8] and astrocytes in the CNS [9]. But again, expression outside of the nervous system was not pursued.
At about the same time that NAALADase was being characterized in the Coyle lab, a novel protein was detected in the LNCaP human prostate cancer cell line by Horoszewicz and colleagues [10]. A few years later, Warren “Skip” Heston’s laboratory at the Cleveland Clinic used Horoszewicz’s 7E-C5 monoclonal antibody to purify the protein from LNCaP cells [11], calling it prostate-specific membrane antigen, or PSMA. After sequencing the purified proteins, they reported that Northern blotting showed no expression outside of the normal and cancerous prostate. A subsequent investigation with Western blotting, however, showed that PSMA was predominantly expressed in the prostate but lower levels were detectable in brain, salivary gland and small intestine [12]. Interestingly, they did not detect the protein in the kidney, even though earlier immunohistochemical reports using the same monoclonal antibody suggested its localization there as well [13]. The intestinal localization made by Heston’s group led to the later characterization of PSMA enzymatic activity as a folate hydrolase, cleaving glutamate from dietary folate polyglutamate to its usable form as folate monoglutamate [14]. At the time, however, no connection to NAALADase was made.
Subsequent immunohistochemistry with improved techniques and more selective antibodies showed a distribution [15,16] that essentially matched the previously reported distribution of NAALADase activity [5]. The expression in human prostate turned out to be species specific; it was later established that dogs, the only other species that spontaneously develops prostate cancer, also expresses PSMA in prostate, with a 5-fold increase in prostate cancer [17]. Eventually, the 7E-C5 monoclonal antibody would be approved for use as a diagnostic for prostate cancer as Prostascint. Unfortunately, since the antibody bound to the intracellular domain of the protein, it was a less than optimal imaging agent for living tissue and targeted therapies that used the antibody for delivery radiotherapy and toxins were not successful [18].
The three threads of the story, PSMA, NAALADase, and FOLH1, only came together once the sequence of the gene coding for the brain enzyme was complete [19-22] and it was finally established that PSMA, NAALADase and FOLH1 were all encoded by the same gene. Still, terminology remained confusing with different names used based on the enzyme’s localization. Part of the confusion in these early years was also due, at least in part, to multiple splice variants and homologs [23,24] as well as a profusion of antibodies with varying affinity between species and tissues. In Figure 1, we show recent immunohistochemical studies from our laboratory that illustrate how dependent localization is depending on the antibody used.
Figure 1.
Throughout the literature there are conflicting reports regarding the tissue expression of PSMA/GCPII protein. Recently, we performed immunohistochemistry using three validated monoclonal PSMA/GCPII antibodies recognizing epitopes in the extracellular domain of PSMA/GCPII: 3E6 (Agilent M362029-2), D7I83 (Cell Signaling 12815) and 1A11 (Kind gift Cyril Barinka). As expected, all three antibodies exhibited strong PSMA/GCPII expression in human prostatic epithelium (brown; A-C). In the kidney however, where PSMA/GCPII is known to be expressed selectively in a subset of renal tubules, striking differences were observed. Strong positive signal in renal tubules was present using 1A11 (stars; D, G), while minimal or no staining was present using 3E6 and D7183 (E,F,H,I). These results illustrate the potential confounding impact of antibody selection on immunohistological outcomes. Scale bars 50 μM in all panels.
In 1992, Slusher joined Zeneca where she and colleagues led the discovery of the first potent GCPII inhibitor, 2-PMPA (IC50=300pM) [25]. Then, in 1999, over a decade after the initial characterization of the brain enzyme, Mike Robinson was a key collaborator with Barbara Slusher and James Vornov on a pivotal study that used 2-PMPA to demonstrate a profound neuroprotective effect following acute ischemic brain injury [26]. This was one of the first steps in establishing the important therapeutic potential of GCPII inhibition in the brain. This report was followed by significant efforts by multiple groups to discover additional GCPII inhibitors, including phosphonic acid-, thiol-, hydroxymate-, and urea-based inhibitors (Figure 2). In parallel with these medicinal chemistry efforts, a number of co-crystal structures were determined for GCPII in complex with various inhibitors [27] providing critical insights into their active site binding modes. For instance, the crystal structures of GCPII in complex with (S)-2-PMPA (PDB code: 2JBJ and 2PVW) revealed that the inhibitor forms a network of interactions with the zinc atoms and key residues in the GCPII active site, resulting in the remarkably high inhibitory potency [28,29]. 2-PMPA and other rationally-designed inhibitors were subsequently utilized to demonstrate the robust therapeutic effects of GCPII inhibition in over 40 animal models of diseases in which hyper-glutamatergic neurotransmission was implicated including pain, drug abuse, epilepsy and cognitive deficits (for review see [30,27]).
Figure 2:
Structures of prototypical GCPII inhibitors and their orally-bioavailable prodrugs.
In the early 2000’s, when both Slusher and Vornov worked at Guilford Pharmaceuticals, they led the early stage clinical testing of a thiol-based GCPII inhibitor termed GPI5693 (2-MPPA) for CNS disorders [31]. Unfortunately, the compound never reached a proof of concept trial in human disease because of immunogenicity findings in GLP animal toxicity studies likely related to the compound’s thiol-containing structure, not its pharmacologic activity. The Slusher laboratory has recently reported efforts to synthesize non-thiol orally bioavailable prodrugs of GCPII inhibitors that could finally allow clinical testing in CNS disorders (Figure 2) [32,33]. They have also reported on intranasal administration of GCPII inhibitors to enhance brain penetration [34,35]. However, as of now, the highly polar nature of the known potent GCPII inhibitors has prevented their clinical development for CNS disorders. In contrast, these inhibitors have emerged with significant diagnostic and therapeutic potential in both cancer and inflammatory diseases.
PSMA Theranostics based on GCPII Binding in Prostate Cancer
Since the original reports of PSMA as a marker for prostate cancer, it has been broadly confirmed that PSMA is highly upregulated (10-100 fold) in the majority of prostate cancer lesions [36,37]. PSMA surface expression correlates with disease progression, androgen independence, and metastasis [36,37]. The association of enhanced PSMA expression to increasing cancer grade defined by Gleason Score has fostered a strong interest in PSMA as a diagnostic and therapeutic target.
In recent years, PSMA imaging with small molecule PET ligands has been shown to be a clinically useful tool for diagnosing and staging prostate cancer [38,39,36,40,41]. As discussed below, these ligands have provided a broader window into changing expression of the enzyme in cancer, inflammation and other pathological processes. Examples of PET imaging using a PSMA-ligand in shown in Figure 3.
Figure 3.
The effect of direct inhibition of PSMA binding by co-administration of 2-PMPA and tracer is shown in mice bearing PSMA -expressing 22Rv1 tumor xenografts. A. In mice given just the tracer, the kidneys, bladder and tumor are clearly shown. B. With the blocking agent, kidney and tumor uptake are reduced. K, kidney; B, bladder, T, tumor. C. Ex vivo quantification of distribution in the model showing direct pharmacologic inhibition results in decrease in normal tissue and tumor (N = 3). Data courtesy of Dr. Daniel Thorek. D. A representative human image of the same tracer in a patient with widely metastatic prostate cancer. The salivary glands and kidney are clearly seen in addition to the tumor uptake. Image courtesy of Dr. Steven Rowe.
In addition to diagnostic utility, PSMA targeted radiotherapy with small molecule ligands has emerged as an extremely promising therapy for metastatic prostate cancer that no longer responds to hormonal therapy, so called metatstatic castration-resistant prostate cancer (mCRPC) [42-44] Because ligand binding to PSMA induces rapid internalization, targeting PSMA actually has the potential to promote accumulation of therapeutic ligands in cancer cells [36,37]. Small molecule PSMA ligands are currently in experimental mCRPC clinical studies including beta therapy (e.g. 177Lu-PSMA-617, 131I-MIP-1095), and alpha therapy (e.g. 225Ac-PSMA-617) [45,46]. Some of these molecules, such as PSMA-617, contain a chelator functional group that enables labeling with isotopes for use as both imaging and therapeutic agents [46,39]. For example, 177Lu-PSMA-617 has been administered to hundreds of late stage mCRPC patients [39,47,48] as a diagnostic. Phase 2 results from a trial of 177Lu-PSMA-617 (ACTRN12615000912583) [49], showed promising results even in a heavily pre-treated patient population who had exhausted benefit from approved therapies [47,48]. Dozens of randomized clinical trials are now registered to confirm the utility and efficacy of PSMA imaging and radiotherapy, with 177Lu-PSMA-617 having recently entered Phase 3 trials in mCRPC ( NCT03511664). These PSMA ligands promise to provide a theranostic approach to mCRPC in which a ligand is used to localize and treat widely metastatic disease in patients with few other options.
Despite significant progress, clinical development of PSMA-targeted agents has been complicated by the physiologic expression of PSMA in normal tissues as would be expected from the history outlined above. In dosimetry studies, exposure of salivary glands and kidney are dose limiting [37,39]. In these clinical studies, PSMA radiotherapeutics have resulted in transient or permanent dry mouth (xerostomia) from salivary gland toxicity, particularly in a recent trial of alpha radionuclides [50,43]. For beta emitters such as 177Lu-PSMA-617, kidney exposure is dose-limiting, restricting treatment of the prostate cancer [51,52,48,53-56,36,37]. While some renal injury might be acceptable in heavily pretreated, late stage patients with widely metastatic disease, the risk of chronic injury prevents use of radiation treatment at early stages of mCRPC [44,49,57].
GCPII inhibitor prodrugs that preferentially shield kidneys and salivary glands
Our recent work has provided evidence that it may be possible to selectively block binding of PSMA-targeted radiotherapy to normal tissue without affecting tumor exposure. An early attempt to mitigate healthy tissue exposure using the highly selective PSMA inhibitor 2-PMPA (IC50=0.3nM) showed that while the dose to the kidneys could be reduced by 83% and nephrotoxicity reduced by 3 months, the exposure also inhibited tumor uptake of the radiotherapeutic by more than 50%. This accelerated tumor growth and significantly reduced overall survival relative to mice that received the radiotherapeutic alone [55]. An example of PSMA block both normal tissue and tumor tracer uptake is shown in Figure 3. Similar results were obtained when 2-PMPA was paired with 131I-MIP-1095 [58]. These results did provide some evidence that tumor and normal tissue could show differential binding, making a shielding approach possible if selectivity could be improved by modification of the shielding ligand to block normal tissue more effectively than the tumor.
We have recently synthesized a tri-alkoxycarbonyloxy alkyl (TrisPOC) prodrug of 2-PMPA (JHU-2545) that appears to preferentially deliver 2-PMPA to non-malignant tissues such as salivary glands and kidneys versus tumor in rodent models. JHU-2545 achieved a 3- and 53-fold enhanced delivery of 2-PMPA to salivary glands and kidneys, respectively, compared to prostate cancer xenografts. We have shown that JHU-2545 can block uptake in rodent salivary glands and kidneys of PSMA targeted radioligands by up to 85% without effect on prostate cancer xenograft uptake and a patent has been published (Slusher et al. PCT/US2018/027106). If similar preferential delivery can be achieved in patients, JHU-2545 or a similar compound could provide a way to bring PSMA-targeted radiotherapy to a wider range of patients with a broader range of beta and alpha emitting radionuclides.
GCPII Inhibitors to Treat Cancer
Since highly specific and potent GCPII inhibitors have been synthesized, an obvious question is whether enzymatic inhibition might also have therapeutic benefit. One idea is that GCPII provides metabolic substrates for cancer growth. It seems possible that PSMA might not only allow increased supply of folate from polyglutamated folate, but also provide a supply of nitrogen, energy and carbon precursors via glutamate and conversion to glutamine [59]. Recently, a correlation between NAAG concentration and cancer grade has been reported [60], with the observation that inhibition of GCPII and glutaminase had synergistic effects on inhibiting tumor growth, supporting the idea that GCPII plays a metabolic role in cancer.
From 1996 to 1999, Slusher and colleagues published several US patents claiming both diagnostic potential and therapeutic utility of GCPII inhibition for cancer (e.g. Slusher et al., US5804602A US5981209A, US6011021A, US6372726B1). Recently, this therapeutic finding was replicated in a publication from Kaittanis and colleagues that provided additional evidence that the somewhat modest effects of GCPII inhibition on tumor growth were due to blockade of glutamate release from a substrate, presumably folate polyglutamate, and subsequent binding to metabotropic glutamate receptors, activating the IP3K/AKT/mTOR pathway [61]. Interestingly, while there are some effects on cell growth from enzyme inhibition, these are much stronger when combined with androgen receptor blockade.
GCPII over-expression in cancer neovasculature
Soon after the original development of the PSMA antibodies, evidence began to accumulate that GCPII is broadly but not universally expressed in tumor vasculature [62,63,16]. For example, in a survey of 275 samples of non-small cell lung cancer, tumor expression was as low as 6% while neovascular expression was found in 49% of cases [64]. Expression was found in 19.4% of a sample of 779 sarcoma cases, with expression increased in more malignant tumors [65].
PSMA enzymatic activity appears to play a role in tumor angiogenesis, as impairments in tumor angiogenesis are observed in PSMA knock out animals and following PSMA inhibitor administration [66]. Recently, a relationship has been established between matrix metalloproteinase 2 (MMP2) and PSMA/GCPII [67]. Sequential proteolysis of laminin (a component of the extracellular matrix) by MMP2 and PSMA/GCPII produced small peptide fragments that increased the rate of endothelial cell migration [68]. This two-step degradation pathway highlights the possible role that PSMA/GCPII activation has on the induction of angiogenesis and metastasis and a potential explanation for its upregulation in cancer neovasculature. However, the impairments in knockout animals and effects of inhibitors do not appear to be powerful enough to make enzyme inhibition a clinically useful chemotherapy, suggesting that the enzyme may be most important as a means of broadly targeting malignancies with toxins or radionuclides as in prostate cancer.
As 68Ga-PSMA-PET imaging has gained favor as a method for staging and restaging of patients with confirmed prostate cancer, binding has been found, as might be expected, in malignant lesions other than prostate such as renal cell carcinoma, breast cancer, bronchial carcinoma and thyroid carcinoma. The literature has recently been reviewed and in general has confirmed that the binding is often due to expression in tumor neovasculature [69]. In Table 1, some examples of tumor uptake are listed to illustrate the potential broad utility of PSMA-targeted PET ligands for imaging tumors over-expressing the enzyme.
Table 1.
Examples of PSMA detection in neoplasms and benign conditions
Examples | Method of detection | |
---|---|---|
Neoplasm | ||
Breast | Breast cancer[104] | Immunohistochemistry |
Ductal carcinoma[105] | 68Ga PSMA-PET/CT | |
Lobular carcinoma[106] | Immunohistochemistry | |
Triple negative breast cancer[107] | 68Ga PSMA-PET/CT | |
Thyroid | Papillary thyroid carcinoma[108] | 68Ga-PSMA-HBED-CC PET/CT |
Follicular thyroid carcinoma[109] | 68Ga-PSMA PET/CT | |
Differentiate thyroid cancer[110] | 68Ga-PSMA-HBED-CC PET/CT | |
Pancreas | Pancreatic cancer[111] | Immunohistochemistry |
Lung | Lung cancer[112] | Immunohistochemistry |
Neurogenic tumors | Peripheral nerve sheath tumor[113] | 68Ga-PSMA PET/CT |
Paravertebral schwannoma[114] | 68Ga-PSMA PET/CT | |
MPNST, undifferentiated sarcoma, rhabdomyosarcoma, neovasculature of synovial sarcoma[115] | Immunohistochemistry | |
Benign | ||
Bone remodeling | Paget Disease[116] | 68Ga PSMA-PET/CT |
Sacral Fracture[117] | 68Ga PSMA-PET/CT | |
Distal radius fracture[118] | 68Ga PSMA-PET/MRI | |
Fibrous dysplasia[119] | 68Ga PSMA-PET/MRI | |
Bone | Tibia plateau, advance osteoarthritis[120] | 68Ga-PSMA PET/CT |
Spleen | Splenic Sarcoidosis[121] | 68Ga PSMA-PET/CT |
Stroke | Subacute stroke[122] | 68Ga-PSMA-HBED-CC PET/CT |
Subacute cerebellar infarction[123] | 68Ga PSMA-PET/CT | |
Misc. | Acrochordon (skin tag)[124] | 68Ga PSMA-PET/MRI |
Neovasculature of nonneoplastic regenerative and reparative tissues[125] | Immunohistochemistry | |
Desmoid tumor[126] | 68Ga-PSMA-HBED-CC PET/CT | |
Cervical, Celiac, and Sacral Ganglia[127] | 68Ga-PSMA-HBED-CC PET/CT | |
Intramuscular myxoma[128] | 68Ga PSMA-PET/CT |
These findings have recently led to the first examples of PSMA being targeted for the treatment of malignancies beyond prostate. In one report, 177Lu-PSMA-617 was administered to a patient with PSMA-positive metastasized leiomyosarcoma [70]. In another Phase II study, the PSMA-targeted cytotoxin conjugate mipsagargin (G-202) resulted in disease stabilization in late stage hepatocellular carcinoma, mediated in part by reducing blood flow to lesions [71].
It should be noted that expression is not limited to tumor neovasculature. In some cases, expression is found in tumors derived from tissues that normally express the enzyme. For example, GCPII is normally expressed in astrocytes [9] and is upregulated in astrocytic tumors. As in prostate cancer, the increased expression is correlated with astrocytic tumor grade [72], In the case of glioblastoma, this has clinically been confirmed with PET imaging [73,74]. Peripheral nervous system structures such as the ganglia of the sympathetic trunk can also be seen on PET images [75], which again is to be expected based on histological findings that GCPII is highly expressed on non-myelinating Schwann cells [76,77] and is increased in tumors derived from Schwann cells such as malignant peripheral nerve sheath tumors [65]. Similarly, cancer cells arising from non-nervous system organs that express GCPII will often also express the enzyme, as reported for renal cell carcinoma [78]. PSMA theranostics may be particularly useful for these tumor types as the enzyme is selectively expressed by malignant cells.
GCPII inhibitors To Treat Inflammatory Bowel Disease
The second human disease outside of the nervous system established to show over-expression of FOLH1 was inflammatory bowel disease. As was the case in prostate cancer, where the link was established through a human cell line, the role of GCPII in IBD was first suggested by study of the human disease, this time by a genome-wide expression investigation [79].
As noted, after PSMA was identified as the gastrointestinal tract folate hydrolase, GCPII’s role in folate absorption was described. GCPII is expressed by epithelial cells of the proximal small intestine [78,16], where it functions to sequentially cleave γ-linked glutamate residues from dietary polyglutamyl folates [14]. This deconjugation to a monoglutamic form is required for folate absorption. Reduced processing of polyglutamyl folates is known to reduce folate bioavailability [80-82]. A naturally occurring mutation in GCPII has been reported, H457Y [83], which correlates with circulating concentrations of folate and its downstream metabolite homocysteine in population studies [83-90]. A detailed mechanistic study highlights the complexity of this relationship, demonstrating that at a structural level the H457Y mutation has no effect on polyglutamyl folate binding or enzyme kinetics, suggesting that an in vivo effect of H475Y polymorphism on folate status is likely to be indirect. [91].
With regard to human disease, multiple independent genome-wide screens have now identified that FOLH1 expression is significantly upregulated in the affected intestinal mucosa of IBD patients [92,93,16,79], where FOLH1 has been described to function as a “hub” gene with significant correlations to over a dozen known IBD gene biomarkers [79]. Correspondingly, it has been validated that GCPII enzymatic activity is consistently and robustly increased in both Crohn’s disease and ulcerative colitis diseased patient biopsies by 300-3000% [94] and that pharmacological inhibition of GCPII ameliorates clinical signs in mouse models [95,94].
GCPII Expression in Other Inflammatory Diseases.
One of the novel observations from the wide use of PSMA PET ligand use in prostate cancer is the expression in benign inflammatory states, including anal fistula [96], sarcoidosis [97], fasciitis [98], and cerebral infarction [99, 100]. PSMA ligand uptake has also been incidentally observed in areas of bone and joint remodeling, such as in Paget disease [101], fractures [102], and synovitis [103], in patients being imaged for their prostate cancer. Table 1 includes some examples of these intriguing incidental findings of increased enzyme expression.
Interestingly, there are no reports of imaging in patients with inflammatory bowel disease to date. There are intriguing suggestions that mGluRs and PI3K/Akt may serve as common active pathogenic signaling systems activated in cancer, inflammatory states and angiogenesis, all influenced by expression of PSMA/GCPII. As the roles of these signaling systems are understood in the pathological states, new approaches to targeted therapy may be possible that address multiple processes across a wide range of diseases, with PSMA PET imaging serving as a means to identify appropriate patients and perhaps useful as a way to assess response to therapy.
Conclusion
Since the first characterization of GCPII enzymatic activity by Mike Robinson and collaborators in the Coyle lab, much has been learned about the enzyme and its function in pathologic conditions in the brain, cancer and inflammatory diseases. The potential therapeutic utility of GCPII is currently most promising using radiochemical ligands as theranostics in cancer and enzyme inhibitors to treat inflammatory bowel disease. While work continues to develop potent brain penetrable GCPII inhibitors, the work started with Mike Robinson many years ago unexpectedly shows the greatest promise to provide treatments for important unmet medical needs outside the nervous system.
Footnotes
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.
References
- 1.Robinson MB, Blakely RD, Coyle JT (1986) Quisqualate selectively inhibits a brain peptidase which cleaves N-acetyl-L-aspartyl-L-glutamate in vitro. Eur J Pharmacol 130 (3):345–347 [DOI] [PubMed] [Google Scholar]
- 2.Coyle JT, Blakely R, Zaczek R, Koller KJ, Abreu M, Ory-Lavollee L, Fisher R, ffrench-Mullen JM, Carpenter DO (1986) Acidic peptides in brain: do they act at putative glutamatergic synapses? Adv Exp Med Biol 203:375–384. doi: 10.1007/978-1-4684-7971-3_28 [DOI] [PubMed] [Google Scholar]
- 3.Ffrench-Mullen JM, Koller K, Zaczek R, Coyle JT, Hori N, Carpenter DO (1985) N-Acetylaspartylglutamate: possible role as the neurotransmitter of the lateral olfactory tract. Proc Natl Acad Sci U S A 82 (11):3897–3900. doi: 10.1073/pnas.82.11.3897 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Blakely RD, Robinson MB, Thompson RC, Coyle JT (1988) Hydrolysis of the brain dipeptide N-acetyl-L-aspartyl-L-glutamate: subcellular and regional distribution, ontogeny, and the effect of lesions on N-acetylated-alpha-linked acidic dipeptidase activity. J Neurochem 50 (4):1200–1209 [DOI] [PubMed] [Google Scholar]
- 5.Robinson MB, Blakely RD, Couto R, Coyle JT (1987) Hydrolysis of the brain dipeptide N-acetyl-L-aspartyl-L-glutamate. Identification and characterization of a novel N-acetylated alpha-linked acidic dipeptidase activity from rat brain. J Biol Chem 262 (30):14498–14506 [PubMed] [Google Scholar]
- 6.Slusher BS, Robinson MB, Tsai GC, Simmons ML, Richards SS, Coyle JT (1990) Rat-Brain N-Acetylated Alpha-Linked Acidic Dipeptidase Activity - Purification and Immunological Characterization. Journal of Biological Chemistry 265 (34):21297–21301 [PubMed] [Google Scholar]
- 7.Slusher BS, Tsai G, Yoo G, Coyle JT (1992) Immunocytochemical Localization of the N-Acetyl-Aspartyl-Glutamate (Naag) Hydrolyzing Enzyme N-Acetylated Alpha-Linked Acidic Dipeptidase (Naaladase). Journal of Comparative Neurology 315 (2):217–229 [DOI] [PubMed] [Google Scholar]
- 8.Berger UV, Carter RE, Mckee M, Coyle JT (1995) N-Acetylated Alpha-Linked Acidic Dipeptidase Is Expressed by Non-Myelinating Schwann-Cells in the Peripheral Nervous-System. Journal of Neurocytology 24 (2):99–109. doi:Doi 10.1007/Bf01181553 [DOI] [PubMed] [Google Scholar]
- 9.Berger UV, Luthi-Carter R, Passani LA, Elkabes S, Black I, Konradi C, Coyle JT (1999) Glutamate carboxypeptidase II is expressed by astrocytes in the adult rat nervous system. Journal of Comparative Neurology 415 (1):52–64. doi:Doi [DOI] [PubMed] [Google Scholar]
- 10.Horoszewicz JS, Kawinski E, Murphy GP (1987) Monoclonal antibodies to a new antigenic marker in epithelial prostatic cells and serum of prostatic cancer patients. Anticancer Res 7 (5B):927–935 [PubMed] [Google Scholar]
- 11.Israeli RS, Powell CT, Fair WR, Heston WD (1993) Molecular cloning of a complementary DNA encoding a prostate-specific membrane antigen. Cancer Res 53 (2):227–230 [PubMed] [Google Scholar]
- 12.Israeli RS, Powell CT, Corr JG, Fair WR, Heston WD (1994) Expression of the prostate-specific membrane antigen. Cancer Res 54 (7):1807–1811 [PubMed] [Google Scholar]
- 13.Wynant GE, Murphy GP, Horoszewicz JS, Neal CE, Collier BD, Mitchell E, Purnell G, Tyson I, Heal A, Abdel-Nabi H, et al. (1991) Immunoscintigraphy of prostatic cancer: preliminary results with 111In-labeled monoclonal antibody 7E11-C5.3 (CYT-356). Prostate 18 (3):229–241 [DOI] [PubMed] [Google Scholar]
- 14.Pinto JT, Suffoletto BP, Berzin TM, Qiao CH, Lin S, Tong WP, May F, Mukherjee B, Heston WD (1996) Prostate-specific membrane antigen: a novel folate hydrolase in human prostatic carcinoma cells. Clin Cancer Res 2 (9):1445–1451 [PubMed] [Google Scholar]
- 15.Troyer JK, Beckett ML, Wright GL Jr., (1995) Detection and characterization of the prostate-specific membrane antigen (PSMA) in tissue extracts and body fluids. Int J Cancer 62 (5):552–558 [DOI] [PubMed] [Google Scholar]
- 16.Silver DA, Pellicer I, Fair WR, Heston WD, Cordon-Cardo C (1997) Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin Cancer Res 3 (1):81–85 [PubMed] [Google Scholar]
- 17.Lai CL, van den Ham R, van Leenders G, van der Lugt J, Mol JA, Teske E (2008) Histopathological and immunohistochemical characterization of canine prostate cancer. Prostate 68 (5):477–488. doi: 10.1002/pros.20720 [DOI] [PubMed] [Google Scholar]
- 18.O'Keefe DS, Bacich DJ, Huang SS, Heston WDW (2018) A Perspective on the Evolving Story of PSMA Biology, PSMA-Based Imaging, and Endoradiotherapeutic Strategies. J Nucl Med 59 (7):1007–1013. doi: 10.2967/jnumed.117.203877 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bzdega T, Turi T, Wroblewska B, She D, Chung HS, Kim H, Neale JH (1997) Molecular cloning of a peptidase against N-acetylaspartylglutamate from a rat hippocampal cDNA library. J Neurochem 69 (6):2270–2277. doi: 10.1046/j.1471-4159.1997.69062270.x [DOI] [PubMed] [Google Scholar]
- 20.Luthi-Carter R, Barczak AK, Speno H, Coyle JT (1998) Hydrolysis of the neuropeptide N-acetylaspartylglutamate (NAAG) by cloned human glutamate carboxypeptidase II. Brain Res 795 (1– 2):341–348. doi: 10.1016/s0006-8993(98)00244-3 [DOI] [PubMed] [Google Scholar]
- 21.Luthi-Carter R, Berger UV, Barczak AK, Enna M, Coyle JT (1998) Isolation and expression of a rat brain cDNA encoding glutamate carboxypeptidase II. Proc Natl Acad Sci U S A 95 (6):3215–3220. doi: 10.1073/pnas.95.6.3215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Carter RE, Feldman AR, Coyle JT (1996) Prostate-specific membrane antigen is a hydrolase with substrate and pharmacologic characteristics of a neuropeptidase. Proc Natl Acad Sci U S A 93 (2):749–753. doi: 10.1073/pnas.93.2.749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hlouchova K, Barinka C, Konvalinka J, Lubkowski J (2009) Structural insight into the evolutionary and pharmacologic homology of glutamate carboxypeptidases II and III. FEBS J 276 (16):4448–4462. doi: 10.1111/j.1742-4658.2009.07152.x [DOI] [PubMed] [Google Scholar]
- 24.Hlouchova K, Navratil V, Tykvart J, Sacha P, Konvalinka J (2012) GCPII Variants, Paralogs and Orthologs. Current Medicinal Chemistry 19 (9):1316–1322. doi: 10.2174/092986712799462676 [DOI] [PubMed] [Google Scholar]
- 25.Jackson PF, Cole DC, Slusher BS, Stetz SL, Ross LE, Donzanti BA, Trainor DA (1996) Design, synthesis, and biological activity of a potent inhibitor of the neuropeptidase N-acetylated alpha-linked acidic dipeptidase. Journal of medicinal chemistry 39 (2):619–622. doi: 10.1021/jm950801q [DOI] [PubMed] [Google Scholar]
- 26.Slusher BS, Vornov JJ, Thomas AG, Hurn PD, Harukuni I, Bhardwaj A, Traystman RJ, Robinson MB, Britton P, Lu XC, Tortella FC, Wozniak KM, Yudkoff M, Potter BM, Jackson PF (1999) Selective inhibition of NAALADase, which converts NAAG to glutamate, reduces ischemic brain injury. Nat Med 5 (12):1396–1402. doi: 10.1038/70971 [DOI] [PubMed] [Google Scholar]
- 27.Barinka C, Rojas C, Slusher B, Pomper M (2012) Glutamate carboxypeptidase II in diagnosis and treatment of neurologic disorders and prostate cancer. Curr Med Chem 19 (6):856–870. doi: 10.2174/092986712799034888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Barinka C, Starkova J, Konvalinka J, Lubkowski J (2007) A high-resolution structure of ligand-free human glutamate carboxypeptidase II. Acta Crystallogr Sect F Struct Biol Cryst Commun 63 (Pt 3):150–153. doi: 10.1107/S174430910700379X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mesters JR, Henning K, Hilgenfeld R (2007) Human glutamate carboxypeptidase II inhibition: structures of GCPII in complex with two potent inhibitors, quisqualate and 2-PMPA. Acta Crystallogr D Biol Crystallogr 63 (Pt 4):508–513. doi: 10.1107/S090744490700902X [DOI] [PubMed] [Google Scholar]
- 30.Vornov JJ, Hollinger KR, Jackson PF, Wozniak KM, Farah MH, Majer P, Rais R, Slusher BS (2016) Still NAAG'ing After All These Years: The Continuing Pursuit of GCPII Inhibitors. Adv Pharmacol 76:215–255. doi: 10.1016/bs.apha.2016.01.007 [DOI] [PubMed] [Google Scholar]
- 31.van der Post JP, de Visser SJ, de Kam ML, Woelfler M, Hilt DC, Vornov J, Burak ES, Bortey E, Slusher BS, Limsakun T, Cohen AF, van Gerven JM (2005) The central nervous system effects, pharmacokinetics and safety of the NAALADase-inhibitor GPI 5693. Br J Clin Pharmacol 60 (2):128–136. doi: 10.1111/j.1365-2125.2005.02396.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Nedelcovych M, Dash RP, Tenora L, Zimmermann SC, Gadiano AJ, Garrett C, Alt J, Hollinger KR, Pommier E, Jancarik A, Rojas C, Thomas AG, Wu Y, Wozniak K, Majer P, Slusher BS, Rais R (2017) Enhanced Brain Delivery of 2-(Phosphonomethyl)pentanedioic Acid Following Intranasal Administration of Its gamma-Substituted Ester Prodrugs. Molecular pharmaceutics 14 (10):3248–3257. doi: 10.1021/acs.molpharmaceut.7b00231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Dash RP, Tichý T, Veeravalli V, Lam J, Alt J, Wu Y, Tenora L, Majer P, Slusher BS, Rais R (2019) Enhanced Oral Bioavailability of 2-(Phosphonomethyl)-pentanedioic Acid (2-PMPA) from its (5-Methyl-2-oxo-1,3-dioxol-4-yl)methyl (ODOL)-Based Prodrugs. Molecular Pharmaceutics 16 (10):4292–4301. doi: 10.1021/acs.molpharmaceut.9b00637 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rais R, Wozniak K, Wu Y, Niwa M, Stathis M, Alt J, Giroux M, Sawa A, Rojas C, Slusher BS (2015) Selective CNS Uptake of the GCP-II Inhibitor 2-PMPA following Intranasal Administration. PLoS One 10 (7):e0131861. doi: 10.1371/journal.pone.0131861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Nedelcovych M, Dash RP, Tenora L, Zimmermann SC, Gadiano AJ, Garrett C, Alt J, Hollinger KR, Pommier E, Jančařík A, Rojas C, Thomas AG, Wu Y, Wozniak K, Majer P, Slusher BS, Rais R (2017) Enhanced Brain Delivery of 2-(Phosphonomethyl)pentanedioic Acid Following Intranasal Administration of Its γ-Substituted Ester Prodrugs. Molecular pharmaceutics 14 (10):3248–3257. doi: 10.1021/acs.molpharmaceut.7b00231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bouchelouche K, Turkbey B, Choyke PL (2016) PSMA PET and Radionuclide Therapy in Prostate Cancer. Seminars in nuclear medicine 46 (6):522–535. doi: 10.1053/j.semnuclmed.2016.07.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ristau BT, O'Keefe DS, Bacich DJ (2014) The prostate-specific membrane antigen: lessons and current clinical implications from 20 years of research. Urologic oncology 32 (3):272–279. doi: 10.1016/j.urolonc.2013.09.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Afshar-Oromieh A, Malcher A, Eder M, Eisenhut M, Linhart HG, Hadaschik BA, Holland-Letz T, Giesel FL, Kratochwil C, Haufe S, Haberkorn U, Zechmann CM (2013) PET imaging with a [68Ga]gallium-labelled PSMA ligand for the diagnosis of prostate cancer: biodistribution in humans and first evaluation of tumour lesions. EurJ Nucl Med Mol Imaging 40 (4):486–495. doi: 10.1007/s00259-012-2298-2 [DOI] [PubMed] [Google Scholar]
- 39.Afshar-Oromieh A, Babich JW, Kratochwil C, Giesel FL, Eisenhut M, Kopka K, Haberkorn U (2016) The Rise of PSMA Ligands for Diagnosis and Therapy of Prostate Cancer. J Nucl Med 57 (Suppl 3):79s–89s. doi: 10.2967/jnumed.115.170720 [DOI] [PubMed] [Google Scholar]
- 40.Sawicki LM, Kirchner J, Buddensieck C, Antke C, Ullrich T, Schimmoller L, Boos J, Schleich C, Schaarschmidt BM, Buchbender C, Heusch P, Rabenalt R, Albers P, Antoch G, Muller HW, Hautzel H (2019) Prospective comparison of whole-body MRI and (68)Ga-PSMA PET/CT for the detection of biochemical recurrence of prostate cancer after radical prostatectomy. Eur J Nucl Med Mol Imaging 46 (7):1542–1550. doi: 10.1007/s00259-019-04308-5 [DOI] [PubMed] [Google Scholar]
- 41.Uprimny C (2019) (68)Ga-PSMA-11 PET/CT: the rising star of nuclear medicine in prostate cancer imaging? Wien Med Wochenschr 169 (l-2):3–ll. doi: 10.1007/sl0354-017-0569-z [DOI] [PubMed] [Google Scholar]
- 42.Fendler WP, Rahbar K, Herrmann K, Kratochwil C, Eiber M (2017) (177)Lu-PSMA Radioligand Therapy for Prostate Cancer. J Nucl Med 58 (8):1196–1200. doi: 10.2967/jnumed.117.191023 [DOI] [PubMed] [Google Scholar]
- 43.Kratochwil C, Bruchertseifer F, Rathke H, Hohenfellner M, Giesel FL, Haberkorn U, Morgenstern A (2018) Targeted alpha-Therapy of Metastatic Castration-Resistant Prostate Cancer with (225)Ac-PSMA-617: Swimmer-Plot Analysis Suggests Efficacy Regarding Duration of Tumor Control. J Nucl Med 59 (5):795–802. doi: 10.2967/jnumed.117.203539 [DOI] [PubMed] [Google Scholar]
- 44.von Eyben FE, Roviello G, Kiljunen T, Uprimny C, Virgolini I, Kairemo K, Joensuu T (2017) Third-line treatment and (177)Lu-PSMA radioligand therapy of metastatic castration-resistant prostate cancer: a systematic review. Eur J Nucl Med Mol Imaging. doi: 10.1007/s00259-017-3895-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Eiber M, Fendler WP, Rowe SP, Calais J, Hofman MS, Maurer T, Schwarzenboeck SM, Kratowchil C, Herrmann K, Giesel FL (2017) Prostate-Specific Membrane Antigen Ligands for Imaging and Therapy. J Nucl Med 58 (Suppl 2):67s–76s. doi: 10.2967/jnumed.116.186767 [DOI] [PubMed] [Google Scholar]
- 46.Virgolini I, Decristoforo C, Haug A, Fanti S, Uprimny C (2018) Current status of theranostics in prostate cancer. Eur J Nucl Med Mol Imaging 45 (3):471–495. doi: 10.1007/s00259-017-3882-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kratochwil C, Giesel FL, Stefanova M, Benesova M, Bronzel M, Afshar-Oromieh A, Mier W, Eder M, Kopka K, Haberkorn U (2016) PSMA-Targeted Radionuclide Therapy of Metastatic Castration-Resistant Prostate Cancer with 177Lu-Labeled PSMA-617. J Nucl Med 57 (8):1170–1176. doi: 10.2967/jnumed.115.171397 [DOI] [PubMed] [Google Scholar]
- 48.Rahbar K, Ahmadzadehfar H, Kratochwil C, Haberkorn U, Schafers M, Essler M, Baum RP, Kulkarni HR, Schmidt M, Drzezga A, Bartenstein P, Pfestroff A, Luster M, Lutzen U, Marx M, Prasad V, Brenner W, Heinzel A, Mottaghy FM, Ruf J, Meyer PT, Heuschkel M, Eveslage M, Bogemann M, Fendler WP, Krause BJ (2017) German Multicenter Study Investigating 177Lu-PSMA-617 Radioligand Therapy in Advanced Prostate Cancer Patients. J Nucl Med 58 (1):85–90. doi: 10.2967/jnumed.116.183194 [DOI] [PubMed] [Google Scholar]
- 49.Hofman MS, Violet J, Hicks RJ, Ferdinandus J, Ping Thang S, Akhurst T, Iravani A, Kong G, Ravi Kumar A, Murphy DG, Eu P, Jackson P, Scalzo M, Williams SG, Sandhu S (2018) [(177)Lu]-PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): a single-centre, single-arm, phase 2 study. Lancet Oncol. doi: 10.1016/s1470-2045(18)30198-0 [DOI] [PubMed] [Google Scholar]
- 50.Taieb D, Foletti JM, Bardies M, Rocchi P, Hicks R, Haberkorn U (2018) PSMA-Targeted Radionuclide Therapy and salivary gland toxicity: why does it matter? J Nucl Med. doi: 10.2967/jnumed.118.207993 [DOI] [PubMed] [Google Scholar]
- 51.Kabasakal L, AbuQbeitah M, Aygun A, Yeyin N, Ocak M, Demirci E, Toklu T (2015) Pre-therapeutic dosimetry of normal organs and tissues of (177)Lu-PSMA-617 prostate-specific membrane antigen (PSMA) inhibitor in patients with castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 42 (13):1976–1983. doi: 10.1007/s00259-015-3125-3 [DOI] [PubMed] [Google Scholar]
- 52.Delker A, Fendler WP, Kratochwil C, Brunegraf A, Gosewisch A, Gildehaus FJ, Tritschler S, Stief CG, Kopka K, Haberkorn U, Bartenstein P, Boning G (2016) Dosimetry for (177)Lu-DKFZ-PSMA-617: a new radiopharmaceutical for the treatment of metastatic prostate cancer. Eur J Nucl Med Mol Imaging 43 (1):42–51. doi: 10.1007/s00259-015-3174-7 [DOI] [PubMed] [Google Scholar]
- 53.Zechmann CM, Afshar-Oromieh A, Armor T, Stubbs JB, Mier W, Hadaschik B, Joyal J, Kopka K, Debus J, Babich JW, Haberkorn U (2014) Radiation dosimetry and first therapy results with a (124)I/ (131)I-labeled small molecule (MIP-1095) targeting PSMA for prostate cancer therapy. Eur J Nucl Med Mol Imaging 41 (7):1280–1292. doi: 10.1007/s00259-014-2713-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kratochwil C, Bruchertseifer F, Rathke H, Bronzel M, Apostolidis C, Weichert W, Haberkorn U, Giesel FL, Morgenstern A (2017) Targeted Alpha Therapy of mCRPC with 225Actinium-PSMA-617: Dosimetry estimate and empirical dose finding. J Nucl Med. doi: 10.2967/jnumed.117.191395 [DOI] [PubMed] [Google Scholar]
- 55.Chatalic KL, Heskamp S, Konijnenberg M, Molkenboer-Kuenen JD, Franssen GM, Clahsen-van Groningen MC, Schottelius M, Wester HJ, van Weerden WM, Boerman OC, de Jong M (2016) Towards Personalized Treatment of Prostate Cancer: PSMA I&T, a Promising Prostate-Specific Membrane Antigen-Targeted Theranostic Agent. Theranostics 6 (6):849–861. doi: 10.7150/thno.14744 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Maurer T, Eiber M, Schwaiger M, Gschwend JE (2016) Current use of PSMA-PET in prostate cancer management. Nature reviews Urology 13 (4):226–235. doi: 10.1038/nrurol.2016.26 [DOI] [PubMed] [Google Scholar]
- 57.Kulkarni H, Schuchardt C, SINGH A, Langbein T, Baum R (2018) Early initiation of Lu-177 PSMA radioligand therapy prolongs overall survival in metastatic prostate cancer. Journal of Nuclear Medicine 59 (supplement 1):52929025984 [Google Scholar]
- 58.Kratochwil C, Giesel FL, Leotta K, Eder M, Hoppe-Tich T, Youssoufian H, Kopka K, Babich JW, Haberkorn U (2015) PMPA for nephroprotection in PSMA-targeted radionuclide therapy of prostate cancer. J Nucl Med 56 (2):293–298. doi: 10.2967/jnumed.114.147181 [DOI] [PubMed] [Google Scholar]
- 59.Lemberg KM, Vornov JJ, Rais R, Slusher BS (2018) We're Not "DON" Yet: Optimal Dosing and Prodrug Delivery of 6-Diazo-5-oxo-L-norleucine. Mol Cancer Ther 17 (9):1824–1832. doi: 10.1158/1535-7163.MCT-17-1148 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Nguyen T, Kirsch BJ, Asaka R, Nabi K, Quinones A, Tan J, Antonio MJ, Camelo F, Li T, Nguyen S, Hoang G, Nguyen K, Udupa S, Sazeides C, Shen YA, Elgogary A, Reyes J, Zhao L, Kleensang A, Chaichana KL, Hartung T, Betenbaugh MJ, Marie SK, Jung JG, Wang TL, Gabrielson E, Le A (2019) Uncovering the Role of N-Acetyl-Aspartyl-Glutamate as a Glutamate Reservoir in Cancer. Cell Rep 27 (2):491–501 e496. doi: 10.1016/j.celrep.2019.03.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kaittanis C, Andreou C, Hieronymus H, Mao N, Foss CA, Eiber M, Weirich G, Panchal P, Gopalan A, Zurita J, Achilefu S, Chiosis G, Ponomarev V, Schwaiger M, Carver BS, Pomper MG, Grimm J (2018) Prostate-specific membrane antigen cleavage of vitamin B9 stimulates oncogenic signaling through metabotropic glutamate receptors. J Exp Med 215 (1):159–175. doi: 10.1084/jem.20171052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chang SS, Reuter VE, Heston WD, Bander NH, Grauer LS, Gaudin PB (1999) Five different anti-prostate-specific membrane antigen (PSMA) antibodies confirm PSMA expression in tumor-associated neovasculature. Cancer Res 59 (13):3192–3198 [PubMed] [Google Scholar]
- 63.Liu H, Moy P, Kim S, Xia Y, Rajasekaran A, Navarro V, Knudsen B, Bander NH (1997) Monoclonal antibodies to the extracellular domain of prostate-specific membrane antigen also react with tumor vascular endothelium. Cancer Res 57 (17):3629–3634 [PubMed] [Google Scholar]
- 64.Schmidt LH, Heitkotter B, Schulze AB, Schliemann C, Steinestel K, Trautmann M, Marra A, Hillejan L, Mohr M, Evers G, Wardelmann E, Rahbar K, Gorlich D, Lenz G, Berdel WE, Hartmann W, Wiewrodt R, Huss S (2017) Prostate specific membrane antigen (PSMA) expression in non-small cell lung cancer. PLoS One 12 (10):e0186280. doi: 10.1371/journal.pone.0186280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Heitkotter B, Trautmann M, Grunewald I, Bogemann M, Rahbar K, Gevensleben H, Wardelmann E, Hartmann W, Steinestel K, Huss S (2017) Expression of PSMA in tumor neovasculature of high grade sarcomas including synovial sarcoma, rhabdomyosarcoma, undifferentiated sarcoma and MPNST. Oncotarget 8 (3):4268–4276. doi: 10.18632/oncotarget.13994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Conway RE, Petrovic N, Li Z, Heston W, Wu D, Shapiro LH (2006) Prostate-specific membrane antigen regulates angiogenesis by modulating integrin signal transduction. Mol Cell Biol 26 (14):5310–5324. doi: 10.1128/MCB.00084-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Conway RE, Joiner K, Patterson A, Bourgeois D, Rampp R, Hannah BC, McReynolds S, Elder JM, Gilfilen H, Shapiro LH (2013) Prostate specific membrane antigen produces pro-angiogenic laminin peptides downstream of matrix metalloprotease-2. Angiogenesis 16 (4):847–860. doi: 10.1007/s10456-013-9360-y [DOI] [PubMed] [Google Scholar]
- 68.Conway RE, Rojas C, Alt J, Novakova Z, Richardson SM, Rodrick TC, Fuentes JL, Richardson NH, Attalla J, Stewart S, Fahmy B, Barinka C, Ghosh M, Shapiro LH, Slusher BS (2016) Prostate-specific membrane antigen (PSMA)-mediated laminin proteolysis generates a pro-angiogenic peptide. Angiogenesis 19 (4):487–500. doi: 10.1007/s10456-016-9521-x [DOI] [PubMed] [Google Scholar]
- 69.Backhaus P, Noto B, Avramovic N, Grubert LS, Huss S, Bogemann M, Stegger L, Weckesser M, Schafers M, Rahbar K (2018) Targeting PSMA by radioligands in non-prostate disease-current status and future perspectives. Eur J Nucl Med Mol Imaging 45 (5):860–877. doi: 10.1007/s00259-017-3922-y [DOI] [PubMed] [Google Scholar]
- 70.Juptner M, Marx M, Zuhayra M, Lutzen U (2019) Experimental 177Lu-PSMA-617 radioligand therapy in a patient with extended metastasized leiomyosarcoma. Nuklearmedizin. doi: 10.1055/a-0914-2486 [DOI] [PubMed] [Google Scholar]
- 71.Mahalingam D, Peguero J, Cen P, Arora SP, Sarantopoulos J, Rowe J, Allgood V, Tubb B, Campos L (2019) A Phase II, Multicenter, Single-Arm Study of Mipsagargin (G-202) as a Second-Line Therapy Following Sorafenib for Adult Patients with Progressive Advanced Hepatocellular Carcinoma. Cancers (Basel) 11 (6). doi: 10.3390/cancers11060833 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Nomura N, Pastorino S, Jiang P, Lambert G, Crawford JR, Gymnopoulos M, Piccioni D, Juarez T, Pingle SC, Makale M, Kesari S (2014) Prostate specific membrane antigen (PSMA) expression in primary gliomas and breast cancer brain metastases. Cancer Cell Int 14 (1):26. doi: 10.1186/1475-2867-14-26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pillai MRA, Nanabala R, Joy A, Sasikumar A, Russ Knapp FF (2016) Radiolabeled enzyme inhibitors and binding agents targeting PSMA: Effective theranostic tools for imaging and therapy of prostate cancer. Nucl Med Biol 43 (11):692–720. doi: 10.1016/j.nucmedbio.2016.08.006 [DOI] [PubMed] [Google Scholar]
- 74.Schwenck J, Tabatabai G, Skardelly M, Reischl G, Beschorner R, Pichler B, la Fougere C (2015) In vivo visualization of prostate-specific membrane antigen in glioblastoma. Eur J Nucl Med Mol I 42 (1):170–171. doi: 10.1007/s00259-014-2921-5 [DOI] [PubMed] [Google Scholar]
- 75.Rischpler C, Beck TI, Okamoto S, Schlitter AM, Knorr K, Schwaiger M, Gschwend J, Maurer T, Meyer PT, Eiber M (2018) (68)Ga-PSMA-HBED-CC Uptake in Cervical, Celiac, and Sacral Ganglia as an Important Pitfall in Prostate Cancer PET Imaging. J Nucl Med 59 (9):1406–1411. doi: 10.2967/jnumed.117.204677 [DOI] [PubMed] [Google Scholar]
- 76.Berger UV, Carter RE, McKee M, Coyle JT (1995) N-acetylated alpha-linked acidic dipeptidase is expressed by non-myelinating Schwann cells in the peripheral nervous system. J Neurocytol 24 (2):99–109 [DOI] [PubMed] [Google Scholar]
- 77.Ebenezer GJ, McArthur JC, Thomas D, Murinson B, Hauer P, Polydefkis M, Griffin JW (2007) Denervation of skin in neuropathies: the sequence of axonal and Schwann cell changes in skin biopsies. Brain 130 (Pt 10):2703–2714. doi: 10.1093/brain/awm199 [DOI] [PubMed] [Google Scholar]
- 78.Kinoshita Y, Kuratsukuri K, Landas S, Imaida K, Rovito PM Jr., Wang CY, Haas GP (2006) Expression of prostate-specific membrane antigen in normal and malignant human tissues. World J Surg 30 (4):628–636. doi: 10.1007/s00268-005-0544-5 [DOI] [PubMed] [Google Scholar]
- 79.Zhang T, Song B, Zhu W, Xu X, Gong QQ, Morando C, Dassopoulos T, Newberry RD, Hunt SR, Li E (2012) An ileal Crohn's disease gene signature based on whole human genome expression profiles of disease unaffected ileal mucosal biopsies. PLoS One 7 (5):e37139. doi: 10.1371/journal.pone.0037139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Halsted CH, Baugh CM, Butterworth CE Jr., (1975) Jejunal perfusion of simple and conjugated folates in man. Gastroenterology 68 (2):261–269 [PubMed] [Google Scholar]
- 81.Halsted CH, Reisenauer AM, Shane B, Tamura T (1978) Availability of monoglutamyl and polyglutamyl folates in normal subjects and in patients with coeliac sprue. Gut 19 (10):886–891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hoffbrand AV, Douglas AP, Fry L, Stewart JS (1970) Malabsorption of dietary folate (Pteroylpolyglutamates) in adult coeliac disease and dermatitis herpetiformis. Br Med J 4 (5727):85–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Devlin AM, Ling EH, Peerson JM, Fernando S, Clarke R, Smith AD, Halsted CH (2000) Glutamate carboxypeptidase II: a polymorphism associated with lower levels of serum folate and hyperhomocysteinemia. Hum Mol Genet 9 (19):2837–2844 [DOI] [PubMed] [Google Scholar]
- 84.Lievers KJ, Kluijtmans LA, Boers GH, Verhoef P, den Heijer M, Trijbels FJ, Blom HJ (2002) Influence of a glutamate carboxypeptidase II (GCPII) polymorphism (1561C-->T) on plasma homocysteine, folate and vitamin B(12) levels and its relationship to cardiovascular disease risk. Atherosclerosis 164 (2):269–273 [DOI] [PubMed] [Google Scholar]
- 85.Vargas-Martinez C, Ordovas JM, Wilson PW, Selhub J (2002) The glutamate carboxypeptidase gene II (C>T) polymorphism does not affect folate status in the Framingham Offspring cohort. J Nutr 132 (6):1176–1179. doi: 10.1093/jn/132.6.1176 [DOI] [PubMed] [Google Scholar]
- 86.Afman LA, Trijbels FJ, Blom HJ (2003) The H475Y polymorphism in the glutamate carboxypeptidase II gene increases plasma folate without affecting the risk for neural tube defects in humans. J Nutr 133 (1):75–77. doi: 10.1093/jn/133.1.75 [DOI] [PubMed] [Google Scholar]
- 87.Chen J, Kyte C, Valcin M, Chan W, Wetmur JG, Selhub J, Hunter DJ, Ma J (2004) Polymorphisms in the one-carbon metabolic pathway, plasma folate levels and colorectal cancer in a prospective study. Int J Cancer 110 (4):617–620. doi: 10.1002/ijc.20148 [DOI] [PubMed] [Google Scholar]
- 88.Devlin AM, Clarke R, Birks J, Evans JG, Halsted CH (2006) Interactions among polymorphisms in folate-metabolizing genes and serum total homocysteine concentrations in a healthy elderly population. Am J Clin Nutr 83 (3):708–713. doi: 10.1093/ajcn.83.3.708 [DOI] [PubMed] [Google Scholar]
- 89.Halsted CH, Wong DH, Peerson JM, Warden CH, Refsum H, Smith AD, Nygard OK, Ueland PM, Vollset SE, Tell GS (2007) Relations of glutamate carboxypeptidase II (GCPII) polymorphisms to folate and homocysteine concentrations and to scores of cognition, anxiety, and depression in a homogeneous Norwegian population: the Hordaland Homocysteine Study. Am J Clin Nutr 86 (2):514–521. doi: 10.1093/ajcn/86.2.514 [DOI] [PubMed] [Google Scholar]
- 90.Mir MM, Dar JA, Dar NA, Dar MS, Salam I, Lone MM, Chowdary NA (2008) Combined impact of polymorphism of folate metabolism genes; glutamate carboxypeptidase, methylene tetrahydrofolate reductase and methionine synthase reductase on breast cancer susceptibility in kashmiri women. Int J Health Sci (Qassim) 2 (1):3–14 [PMC free article] [PubMed] [Google Scholar]
- 91.Navratil M, Ptacek J, Sacha P, Starkova J, Lubkowski J, Barinka C, Konvalinka J (2014) Structural and biochemical characterization of the folyl-poly-gamma-l-glutamate hydrolyzing activity of human glutamate carboxypeptidase II. FEBS J 281 (14):3228–3242. doi: 10.1111/febs.12857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ben-Shachar S, Yanai H, Baram L, Elad H, Meirovithz E, Ofer A, Brazowski E, Tulchinsky H, Pasmanik-Chor M, Dotan I (2013) Gene expression profiles of ileal inflammatory bowel disease correlate with disease phenotype and advance understanding of its immunopathogenesis. Inflamm Bowel Dis 19 (12):2509–2521. doi: 10.1097/01.MIB.0000437045.26036.00 [DOI] [PubMed] [Google Scholar]
- 93.Noble CL, Abbas AR, Lees CW, Cornelius J, Toy K, Modrusan Z, Clark HF, Arnott ID, Penman ID, Satsangi J, Diehl L (2010) Characterization of intestinal gene expression profiles in Crohn's disease by genome-wide microarray analysis. Inflamm Bowel Dis 16 (10):1717–1728. doi: 10.1002/ibd.21263 [DOI] [PubMed] [Google Scholar]
- 94.Rais R, Jiang W, Zhai H, Wozniak KM, Stathis M, Hollinger KR, Thomas AG, Rojas C, Vornov JJ, Marohn M, Li X, Slusher BS (2016) FOLH1/GCPII is elevated in IBD patients, and its inhibition ameliorates murine IBD abnormalities. JCI Insight 1 (12). doi: 10.1172/jci.insight.88634 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Date AA, Rais R, Babu T, Ortiz J, Kanvinde P, Thomas AG, Zimmermann SC, Gadiano AJ, Halpert G, Slusher BS, Ensign LM (2017) Local enema treatment to inhibit FOLH1/GCPII as a novel therapy for inflammatory bowel disease. J Control Release 263:132–138. doi: 10.1016/j.jconrel.2017.01.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Klingenberg S, Jochumsen MR, Nielsen TF, Bouchelouche K (2019) 68Ga-PSMA Uptake in Anal Fistula on PET/CT Scan. Clin Nucl Med 44 (1):e54–e56. doi: 10.1097/RLU.0000000000002370 [DOI] [PubMed] [Google Scholar]
- 97.Ardies PJ, Gykiere P, Goethals L, De Mey J, De Geeter F, Everaert H (2017) PSMA Uptake in Mediastinal Sarcoidosis. Clin Nucl Med 42 (4):303–305. doi: 10.1097/RLU.0000000000001543 [DOI] [PubMed] [Google Scholar]
- 98.Henninger M, Maurer T, Hacker C, Eiber M (2016) 68Ga-PSMA PET/MR Showing Intense PSMA Uptake in Nodular Fasciitis Mimicking Prostate Cancer Metastasis. Clin Nucl Med 41 (10):e443–444. doi: 10.1097/RLU.0000000000001310 [DOI] [PubMed] [Google Scholar]
- 99.Chan M, Hsiao E (2017) Subacute Cortical Infarct Showing Uptake on 68Ga-PSMA PET/CT. Clin Nucl Med 42 (2):110–111. doi: 10.1097/RLU.0000000000001489 [DOI] [PubMed] [Google Scholar]
- 100.Noto B, Vrachimis A, Schafers M, Stegger L, Rahbar K (2016) Subacute Stroke Mimicking Cerebral Metastasis in 68Ga-PSMA-HBED-CC PET/CT. Clin Nucl Med 41 (10):e449–451. doi: 10.1097/RLU.0000000000001291 [DOI] [PubMed] [Google Scholar]
- 101.Sasikumar A, Joy A, Nanabala R, Pillai MR, T AH (2016) 68Ga-PSMA PET/CT False-Positive Tracer Uptake in Paget Disease. Clin Nucl Med 41 (10):e454–455. doi: 10.1097/RLU.0000000000001340 [DOI] [PubMed] [Google Scholar]
- 102.Panagiotidis E, Paschali A, Giannoula E, Chatzipavlidou V (2019) Rib Fractures Mimicking Bone Metastases in 18F-PSMA-1007 PET/CT for Prostate Cancer. Clin Nucl Med 44 (1):e46–e48. doi: 10.1097/RLU.0000000000002354 [DOI] [PubMed] [Google Scholar]
- 103.Kirchner J, Schaarschmidt BM, Sawicki LM, Heusch P, Hautzel H, Ermert J, Rabenalt R, Antoch G, Buchbender C (2017) Evaluation of Practical Interpretation Hurdles in 68Ga-PSMA PET/CT in 55 Patients: Physiological Tracer Distribution and Incidental Tracer Uptake. Clin Nucl Med 42 (7):e322–e327. doi: 10.1097/RLU.0000000000001672 [DOI] [PubMed] [Google Scholar]
- 104.Kasoha M, Unger C, Solomayer E-F, Bohle RM, Zaharia C, Khreich F, Wagenpfeil S, Juhasz-Böss I (2017) Prostate-specific membrane antigen (PSMA) expression in breast cancer and its metastases. Clinical & experimental metastasis 34 (8):479–490 [DOI] [PubMed] [Google Scholar]
- 105.Kumar R, Mittal BR, Bhattacharya A, Singh H, Singh SK (2018) Synchronous Detection of Male Breast Cancer and Prostatic Cancer in a Patient With Suspected Prostatic Carcinoma on 68Ga-PSMA PET/CT Imaging. Clinical nuclear medicine 43 (6):431–432 [DOI] [PubMed] [Google Scholar]
- 106.Tolkach Y, Gevensleben H, Bundschuh R, Koyun A, Huber D, Kehrer C, Hecking T, Keyver-Paik M-D, Kaiser C, Ahmadzadehfar H (2018) Prostate-specific membrane antigen in breast cancer: a comprehensive evaluation of expression and a case report of radionuclide therapy. Breast cancer research and treatment 169 (3):447–455 [DOI] [PubMed] [Google Scholar]
- 107.Passah A, Arora S, Damle NA, Tripathi M, Bal C, Subudhi TK, Arora G (2018) 68Ga-Prostate-Specific Membrane Antigen PET/CT in Triple-Negative Breast Cancer. Clinical nuclear medicine 43 (6):460–461 [DOI] [PubMed] [Google Scholar]
- 108.Taywade SK, Damle NA, Bal C (2016) PSMA expression in papillary thyroid carcinoma: opening a new horizon in management of thyroid cancer? Clinical nuclear medicine 41 (5):e263–265 [DOI] [PubMed] [Google Scholar]
- 109.Sager S, Vatankulu B, Uslu L, Sönmezoglu K (2016) Incidental detection of follicular thyroid carcinoma in 68Ga-PSMA PET/CT imaging. Journal of nuclear medicine technology 44 (3):199–200 [DOI] [PubMed] [Google Scholar]
- 110.Verburg FA, Krohn T, Heinzel A, Mottaghy FM, Behrendt FF (2015) First evidence of PSMA expression in differentiated thyroid cancer using [Λ sup 68Λ Ga] PSMA-HBED-CC PET/CT. European journal of nuclear medicine and molecular imaging 42 (10):1622. [DOI] [PubMed] [Google Scholar]
- 111.Ren H, Zhang H, Wang X, Liu J, Yuan Z, Hao J (2014) Prostate-specific membrane antigen as a marker of pancreatic cancer cells. Medical Oncology 31 (3):857. [DOI] [PubMed] [Google Scholar]
- 112.Wang H-l, Wang S-s, Song W-h, Pan Y, Yu H-p, Si T-g, Liu Y, Cui X-n, Guo Z (2015) Expression of prostate-specific membrane antigen in lung cancer cells and tumor neovasculature endothelial cells and its clinical significance. PloS one 10 (5):e0125924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Vamadevan S, Le K, Shen L, Ha L, Mansberg R (2017) Incidental Prostate-Specific Membrane Antigen Uptake in a Peripheral Nerve Sheath Tumor. Clinical nuclear medicine 42 (7):560–562 [DOI] [PubMed] [Google Scholar]
- 114.Rischpler C, Maurer T, Schwaiger M, Eiber M (2016) Intense PSMA-expression usingΛ sup 68Λ Ga-PSMA PET/CT in a paravertebral schwannoma mimicking prostate cancer metastasis. European journal of nuclear medicine and molecular imaging 43 (1):193. [DOI] [PubMed] [Google Scholar]
- 115.Heitkötter B, Trautmann M, Grünewald I, Bögemann M, Rahbar K, Gevensleben H, Wardelmann E, Hartmann W, Steinestel K, Huss S (2017) Expression of PSMA in tumor neovasculature of high grade sarcomas including synovial sarcoma, rhabdomyosarcoma, undifferentiated sarcoma and MPNST. Oncotarget 8 (3):4268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Artigas C, Alexiou J, Garcia C, Wimana Z, Otte F, Gil T, Van Velthoven R, Flamen P (2016) Paget bone disease demonstrated on (68) Ga-PSMA ligand PET/CT. Eur J Nucl Med Mol Imaging 43 (1):195–196 [DOI] [PubMed] [Google Scholar]
- 117.Gykiere P, Goethals L, Everaert H (2016) Healing Sacral Fracture Masquerading as Metastatic Bone Disease on a 68Ga-PSMA PET/CT. Clinical nuclear medicine 41 (7):e346–347 [DOI] [PubMed] [Google Scholar]
- 118.Hoberück S, Michler E, Kaiser D, Röhnert A, Zöphel K, Kotzerke J (2018) Prostate-Specific Membrane Antigen Expression in Distal Radius Fracture. Clinical nuclear medicine 43 (8):611–613 [DOI] [PubMed] [Google Scholar]
- 119.De Coster L, Sciot R, Everaerts W, Gheysens O, Verscuren R, Deroose CM, Pans S, Van Laere K, Goffin KE (2017) Fibrous dysplasia mimicking bone metastasis on 68 GA-PSMA PET/MRI. European journal of nuclear medicine and molecular imaging 44 (9):1607–1608 [DOI] [PubMed] [Google Scholar]
- 120.Kirchner J, Schaarschmidt BM, Sawicki LM, Heusch P, Hautzel H, Ermert J, Rabenalt R, Antoch G, Buchbender C (2017) Evaluation of practical interpretation hurdles in 68Ga-PSMA PET/CT in 55 patients: physiological tracer distribution and incidental tracer uptake. Clinical nuclear medicine 42 (7):e322–e327 [DOI] [PubMed] [Google Scholar]
- 121.Kobe C, Maintz D, Fischer T, Drzezga A, Chang D-H (2015) Prostate-Specific Membrane Antigen PET/CT in Splenic Sarcoidosis. Clinical nuclear medicine 40 (11):897–898 [DOI] [PubMed] [Google Scholar]
- 122.Noto B, Vrachimis A, Schäfers M, Stegger L, Rahbar K (2016) Subacute Stroke Mimicking Cerebral Metastasis in 68Ga-PSMA-HBED-CC PET/CT. Clinical nuclear medicine 41 (10):e449–451 [DOI] [PubMed] [Google Scholar]
- 123.Oh G, Miles K (2018) Subacute Cerebellar Infarction With Uptake on 68Ga-Prostate-Specific Membrane Antigen PET/CT. Clinical nuclear medicine 43 (2):134–135 [DOI] [PubMed] [Google Scholar]
- 124.Daglioz GG, Hekimsoy T, Isgoren S, Sikar AA, Demir H (2017) Uptake of an Acrochordon Incidentally Detected on 68Ga Prostate-Specific Membrane Antigen PET/CT. Clinical nuclear medicine 42 (6):461–462 [DOI] [PubMed] [Google Scholar]
- 125.Gordon IO, Tretiakova MS, Noffsinger AE, Hart J, Reuter VE, Al-Ahmadie HA (2008) Prostate-specific membrane antigen expression in regeneration and repair. Modern Pathology 21 (12):1421. [DOI] [PubMed] [Google Scholar]
- 126.Kanthan GL, Hsiao E, Kneebone A, Eade T, Schembri GP (2016) Desmoid Tumor Showing Intense Uptake on 68Ga PSMA-HBED-CC PET/CT. Clinical nuclear medicine 41 (6):508–509 [DOI] [PubMed] [Google Scholar]
- 127.Rischpler C, Beck TI, Okamoto S, Schlitter AM, Knorr K, Schwaiger M, Gschwend J, Maurer T, Meyer PT, Eiber M (2018) 68Ga-PSMA-HBED-CC uptake in cervical, celiac, and sacral ganglia as an important pitfall in prostate cancer PET imaging. Journal of Nuclear Medicine 59 (9):1406–1411 [DOI] [PubMed] [Google Scholar]
- 128.Zacho HD, Nielsen JB, Dettmann K, Hjulskov SH, Petersen LJ (2017) 68Ga-PSMA PET/CT Uptake in Intramuscular Myxoma Imitates Prostate Cancer Metastasis. Clinical nuclear medicine 42 (6):487–488 [DOI] [PubMed] [Google Scholar]