Vasoactive intestinal peptide receptor-based imaging and treatment of tumors (Review)

  • Authors:
    • Bo Tang
    • Xin Yong
    • Rui Xie
    • Qian-Wei Li
    • Shi-Ming Yang
  • View Affiliations

  • Published online on: January 24, 2014     https://doi.org/10.3892/ijo.2014.2276
  • Pages: 1023-1031
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Vasoactive intestinal peptide receptors (VIPRs) are members of the G-protein-coupled receptor superfamily. These receptors are overexpressed in many common malignant tumors and play a major role in the progression and angiogenesis of a number of malignancies. Therefore, VIPRs may be a valuable target for the molecular imaging of tumors and therapeutic interventions. The specific natural ligand or its analogs can be labeled with a radionuclide and used for tumor receptor imaging, which could be used to visualize VIPR-related surface protein expression in vivo and to monitor the in vivo effects of molecular drugs on tumors. Moreover, the involvement of VIPRs in malignant transformation and angiogenesis renders them potential therapeutic targets for cancer treatment. A variety of VIP antagonists and cytotoxic VIP conjugates have been synthesized and evaluated for VIPR-targeted molecular therapy. The importance of VIPRs in tumor biology and the ability to predict responses to targeted therapy and monitor drug interventions suggest that VIP receptor-based imaging and treatment will be critical for the early diagnosis and management of cancer. Here, we review the current literature regarding VIPRs and their natural ligands and the involvement of VIPRs in tumor growth and angiogenesis, with an emphasis on the present use of VIPRs for the molecular imaging of tumors and therapies targeting VIPRs.

Contents

Introduction

Structural and functional features of VIP/PACAP and VIPRs

VIP receptors as potential targets for imaging and therapy

Targeting VIPRs for molecular imaging

123I-labeled VIP for receptor imaging

99mTc-labeled VIP analogs for receptor imaging

PET molecular imaging of the VIP receptor

Targeting VIP receptor for cancer molecular therapy

Prospective and Conclusion

Introduction

Malignant tumors have become one of the most lethal diseases in humans, and the incidence, diagnostic studies and therapeutic options for tumor treatment have undergone major changes in recent years. Currently, the standard treatment for a malignancy includes surgical resection followed by the delivery of chemotherapeutic agents. Although surgical resection is the preferred choice for early-stage solid tumors, it is not a suitable treatment for invasive, metastatic or hematologic malignancies. The overall toxicity and the side effects of chemotherapy also limit the dose regimen, thus requiring a relatively narrow therapeutic index, which can lead to insufficient and/or unpredictable responses. Furthermore, during the course of therapy, multidrug resistance can severely limit the effectiveness of chemotherapy and is also associated with tumor recurrence (14). Therefore, it is desirable to target therapeutic molecules to primary tumors and their metastases, which represents a major challenge for improving current cancer therapy (5). Another significant challenge is the development of strategies that can guide the use of these targeted molecules and enable the specific delivery of therapeutic agents to malignant tissues (6). Targeted therapies generally take advantage of biological molecules that are uniquely expressed or significantly overexpressed in tumors. Hormone receptors were some of the earliest targets utilized for the targeted treatment of many cancers, such as breast, prostate and thyroid cancers (7,8). Therefore, the molecular imaging of the expression of tumor-related receptors is advantageous because it provides information for receptor-targeted therapy (9).

Molecular imaging has been defined as the visualization, characterization and measurement of biological processes at the molecular and cellular levels (10). Tumor receptor imaging is an important type of molecular imaging and can be used to assess receptor expression for the entire disease burden to facilitate early diagnosis, suggest personalized therapy options, and monitor the in vivo effects of a drug on its target. Different receptors are overexpressed in specific tumor types. Several tumor receptors have been utilized for imaging and therapy, including somatostatin receptors, human epidermal growth factor receptor 2 (HER2), integrin receptors, epidermal growth factor receptor (EGFR), and vasoactive intestinal peptide receptors (VIPRs). Vasoactive intestinal peptide receptors (VIPRs) are highly overexpressed in human tumors and metastases. VIPRs also play a major role in the progression of a number of malignancies, thus suggesting that these receptors may serve as molecular targets for cancer diagnosis and treatment. Herein, we describe the biology of VIPRs and VIPR-targeting agents and discuss the potential application of VIPRs for the diagnosis and treatment of cancer.

Structural and functional features of VIP/PACAP and VIPRs

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are two members of a structurally related family of peptides that includes mammalian peptide histidine methionine amide (PHM), secretin, glucagon, and growth hormone-releasing factor (GRF) (11). VIP was isolated from the small intestine of a pig and was characterized in the early 1970s by Mutt and Said. VIP exhibits various physiological effects, including increased vasodilatation and reduced arterial blood pressure (12), smooth muscle relaxation (13), stimulation of electrolyte secretion (14), immunosuppression, hormonal secretion, cell proliferation and increased gastric motility (15,16). The human VIP gene is located on chromosome 6 at band q25 (17,18) and encodes the 170-amino acid precursor protein, prepro-VIP, which undergoes post-translational modification to produce the 28 amino acid VIP peptide (12,19), in addition to the 27 aa peptide histidine methionine (PHM) in humans or the peptide histidine isoleucine (PHI) in rodents (20). These two peptides are co-synthesized with VIP as part of the same precursor peptide, which is encoded by an adjacent exon in the human genome, suggesting that the family of peptides has evolved via exon duplication coupled with gene duplication (21). The N-terminus of VIP plays an important role in this protein’s biological activity, and several studies have shown that the N-terminus is essential for receptor activation; however, it is not involved in the recognition of the receptor-binding site, which seems to involve amino acids in the C-terminal domain (22,23). The human PACAP gene is located on chromosome 18p11 and encodes a 176-amino acid precursor protein (preproPACAP) that contains PACAP and PACAP-related peptide (PRP) in the C-terminal domain and a 24 amino acid signal protein in the N-terminal domain (23,24).

These two peptides share a similar sequence and exert their functions through the binding of vasoactive intestinal peptide receptors (VIPRs), which are functional receptors for VIP and PACAP. VIPRs are members of the G-protein-coupled receptor (GPCR) family, which is comprised of three classes of receptors: VPAC1, VPAC2 and PAC1. VPAC1 and VPAC2 respond to VIP and PACAP with comparable affinity, whereas PAC1 displays a high affinity for PACAP and a low affinity for VIP (2527). The VIPRs share a similar structural organization; they contain seven transmembrane domains (TM), a large N-terminal segment that includes the binding site of VIP/PACAP, and an intracellular C-terminal region that is associated with signal transduction (28). Upon ligand binding, these receptors mediate their signal transduction activity via the binding of heterotrimeric G-proteins to the receptor’s intracellular loop, which ultimately results in cAMP production (29,30), protein kinase A (PKA) pathway activation (25,26), inositol phosphate (IP) turnover (23), the activation of MAPKs (mitogen-activated protein kinases) (31,32), and the activation of the NF-κB pathway (Fig. 1) (33).

The effects of VIP and PACAP are mediated by VIPRs and have many overlapping functions. These peptides play roles in the gastrointestinal, immune, reproductive, respiratory, cardiovascular and endocrine systems (34,35). For example, VIP is involved in gut motility and acts as a potent vasoregulatory hormone, whereas PACAP is associated with the nervous system and exerts a hypophysiotropic effect on pituitary hormone secretion (11,23). In addition to the crucial roles that VIPRs play in many physiological processes, VIPRs are involved in the progression of a number of malignancies and can stimulate tumor growth and angiogenesis through the transactivation of epidermal growth factor receptor (EGFR) and the expression of vascular endothelial growth factor (VEGF) (36,37).

VIP receptors as potential targets for imaging and therapy

VIPRs have been identified in several normal tissues and in various types of tumors. In normal human tissues, VPAC1 receptors are abundant in the brain, T lymphocytes, and most peripheral tissues including the liver, lungs and intestines (38,39). VPAC2 receptors are mainly expressed in the hippocampus, the brainstem, the spinal cord, and most smooth muscle tissues (39,40). PAC1 receptors are predominantly found in the adrenal medulla and on the surface of neuroendocrine neoplasms (41,42). However, VIPRs are highly overexpressed in the majority of human tumors and are particularly common in frequently occurring cancers. Reubi et al (43) reported that VPAC1 receptors are overexpressed in most frequently occurring malignant epithelial neoplasms, such as cancers of the colon, breast, lungs and prostate, in a pattern similar to that found in normal tissues. In contrast to the ubiquitous expression of VPAC1 receptors in most human tumors, tumors that express VPAC2 receptors are rare, and this phenomenon is only observed in some leiomyomas and gastrointestinal stromal tumors (43,44). A high incidence of PAC1 receptor expression was found in neuroendocrine neoplasms located in the brain and the endocrine and neuroendocrine systems and included gliomas, neuroblastomas, and pituitary adenomas, as well as endometrial cancers (43). The VIPRs are significantly overexpressed in most human tumors, and the differences in the cell surface receptor profiles between cancer cells and their normal counterparts can be utilized as a molecular signature for targeted imaging. Thus, the VIPRs may be potential targets for molecular imaging.

It is well documented that excessive VIPR signaling, which arises from receptor overexpression and autocrine stimulation by VIP/PACAP or other factors, is a hallmark of a wide variety of tumors. Previous research has shown that the serum levels of VIP in several tumors are significantly increased, which may serve as an indicator of the presence of certain tumors and could become a part of their diagnosis (45). Since most tumors overexpress VIPRs, VIP/PACAP can bind to these VIPRs and activate them, leading to an interaction with a stimulatory guanine nucleotide binding protein (Gs) and ultimately resulting in the stimulation of adenylate cyclase. Research has shown that the addition of VIP to lung cancer cells elevated the cAMP levels and led to the activation of a series of transcription factors that promoted the expression of nuclear oncogenes and growth factors (46,47). In addition, VIP activation increased the secretion of VEGF and facilitated tumor angiogenesis via the cAMP/PKA and PI3K signaling pathways (36). The involvement of VIPRs in malignant transformation and angiogenesis renders them potentially valuable targets for cancer therapy.

Targeting VIPRs for molecular imaging

Tumor receptor imaging plays an important role in molecular imaging, the characterization of receptor expression, tumor biology, the identification of molecular targets, and the application of targeted cancer therapy (48). The advantages of tumor receptor imaging include its noninvasive nature, the ability to assess receptor expression, and the ability to evaluate the in vivo effects of the drug on the tumor. Several types of imaging agents may be used for the imaging of receptors that are overexpressed in tumors, including antibodies or their fragments, natural peptide ligands or their analogs, and non-peptide small molecules. Although the introduction of antibodies as specific agents to target malignant tumors dates back almost 35 years, these proteins are insufficient for tumor receptor imaging due to their high molecular weight, slow targeting of the tumor, slow clearance from the blood and normal tissues, and relatively low tumor to non-tumor ratios when used in tumor imaging and therapy (49,50). In general, natural ligands and small ligand analogs for targeted imaging are the most common imaging agents in use because small peptide ligands are small and readily diffusible and have rapid kinetics (51). Another important molecular imaging tool is the use of a tracer that accumulates at the sites of targeted receptors. Most molecular imaging tracers include a reporter moiety that emits a signal that can be detected by a special external device and a linker joining the reporter moiety to the targeted ligands. Some of the most commonly used tracers are radionuclide-labeled probes and optical imaging probes. However, the tissue penetration of optical signals is limited to a few millimeters and thus limits the clinical application of optical imaging probes. Therefore, radionuclide-based tracers dominate in clinical molecular imaging techniques due to their high sensitivity and increased tissue penetration. Currently, there are two major radionuclide-based imaging methods used in the pre-clinical or clinical settings: single photon emission computed tomography (SPECT) and positron emission tomography (PET). For the reasons stated above, we focus on radionuclide-labeled peptide ligands and small molecule analogs used for SPECT and PET imaging in this review.

123I-labeled VIP for receptor imaging

Virgolini et al (52) first reported that 123I-labeled VIP could be successfully used in vivo to scan patients with gastrointestinal adenocarcinomas, carcinoids and insulinomas. After the intravenous injection of the 123I-labeled VIP, ∼45% of the radioactivity was found in the lungs within 30 min, and the activity decreased rapidly. Visualization of the primary tumors and metastases was possible within an hour of injection and was still apparent at 24-h post-injection. Most importantly, radiolabeled VIP-receptor scanning is more advantageous than CT scanning because receptor imaging is based on the receptor expression pattern rather than the macroscopic morphology (52). Subsequently, the same group performed a series of clinical studies in which 123I-labeled VIP was used to image various tumors, such as colorectal cancers (53), pancreatic cancers (54), VIPomas (55), and endocrine tumors (56). These imaging results exhibited a high sensitivity, which was an advantage over CT scanning, particularly in patients with tumors of a small size. Raderer et al (57) compared the in vitro and in vivo binding of 123I-VIP and an 111In-labeled monoclonal antibody specific to the TAG-72 protein in patients with intestinal adenocarcinomas in a single-blinded prospectively randomized trial. The results indicate that VIP receptor scanning is more sensitive than immunoscintigraphy for the localization of intestinal adenocarcinomas and metastatic spread (57). Although 123I-VIP has promising potential to localize even small tumors expressing VIPRs and is useful for the early diagnosis and treatment of tumors, naturally occurring peptides like VIP exhibit limitations for in vivo tumor imaging because they have a short biological half-life and are rapidly degraded in the liver and kidneys. Due to this metabolic instability, native VIP peptides require further chemical modifications to improve their bioavailability for receptor binding prior to their use in tumor receptor imaging. In addition, the radionuclide 123I is generated by an expensive cyclotron instrument, which increases the cost of treatment. Furthermore, radiolabeled VIP requires further isolation and purification, which greatly limits its clinical applications. Therefore, novel stable analogs of VIP and applicable radionuclides are required for VIPR imaging.

99mTc-labeled VIP analogs for receptor imaging

In contrast to 123I, 99mTc has excellent physical characteristics for scintigraphic imaging, is very inexpensive, and is produced by a 99Mo generator system (Table I) (58). In an initial attempt to employ 99mTc labeling, the N terminus (His1) of VIP was conjugated to one of two well-known bi-functional chelating agents (BFCAs): CPTA [4-(1,4,8,11-tetraazacyclotetradec-1-yl) methyl] benzoic acid or MAG3[N-[N[N-(benzylthio) acetyl]-glycyl] glycyl] glycine]. These compounds have two major drawbacks: radiochemical impurity and a loss of biological function. It was found that the histidine residue in the number one position of VIP played an important role in the biological activity of VIP (59).

Table I.

Radionuclides for labeling of molecular imaging and therapeutic probes.

Table I.

Radionuclides for labeling of molecular imaging and therapeutic probes.

NuclideHalf-life (h)ProductionEnergy (keV)Application
99mTc6.01Generatorγ 140SPECT imaging
123I13.27Cyclotronγ 159SPECT imaging
18F1.8Cyclotronβ+ 634 γ 511PET imaging
64Cu12.7Cyclotronβ+ 655 β 573PET imaging
111In67Cyclotronγ 171 Auger e- 30SPECT imaging
68Ga1.1Generatorβ+ 1899 γ 511PET imaging
90Y65Generatorβ 2964Therapy
131I192Cyclotronβ+ 607 γ 364Therapy
177Lu161Cyclotronβ 498 γ 208Therapy

To improve the radiolabeling efficiency, Thakur et al (60) further modified VIP at the carboxy terminus of Asn28. The research group chose to use 4-aminobutyric acid (Aba) as the spacer and extended the molecule to include Gly-Gly-(D)-Aba-Gly, which resulted in an N4 configuration that could be used for the chelation of 99mTc (Fig. 2). This modified analog was referred to as TP3654 (VIP labeled with 99mTc). The biological activity of 99mTc-TP3654 was equivalent to that of the native VIP peptide, and the 24-h tumor uptake of 99mTc-TP3654 was higher than that of 123I-VIP. These results implied that a simple and efficient hybrid peptide technique had been developed for labeling peptides with 99mTc. Subsequently, Thakur et al (58,60,61) evaluated the pharmacokinetics and imaging characteristics of 99mTc-TP3654 in normal volunteers and patients with a history of cancer. No adverse reactions or changes were noted in the imaging process. Furthermore, ∼70% of the injected radioactivity was eliminated in the urine, and 20% was cleared through the liver. The tumor uptake of 99mTc-TP3654 was significantly higher than that of 111In-octreotide, which is concordant with the higher VIP receptor density observed in most tumors when compared to the density of somatostatin receptors. This VIP analog promises to be a nontoxic and reliable agent for imaging human tumors that overexpress VIPRs.

Kothari et al (62) synthesized three VIP analogs functionalized at the N terminus with histidine [VP05 (28 residue), VD4 (20 residue), VD5 (19 residue)] by solid phase synthesis and radiolabeled the VIP analogs with 99mTc via a novel tricarbonyl synthon. All of the radiolabeled complexes were stable at room temperature and were produced with high yields. Biodistribution studies revealed good tumor uptake kinetics; however, the soft tissue uptake was also high due to the lipophilic nature of the peptide. Thus, more research is required to modify the reported analogs and create novel radiolabeled analogs to enhance their ability to target VIP receptors.

PET molecular imaging of the VIP receptor

Although PET is not as widely available as SPECT, PET is advantageous for tumor diagnosis because it exhibits higher resolution and sensitivity than computed tomography (CT), magnetic resonance imaging (MRI) and SPECT. Therefore, labeling VIP with positron emission radionuclides is important for PET molecular imaging of tumors expressing VIP receptors. Moody et al (63) used 18F to radiolabel VIP analogs (at Arg15/Arg21) and obtained a labeled complex (18F-RR) VIP that bound to breast cancer cells with high specificity and affinity and acted as an agonist. The tumor imaging results showed that 4 h after injection, the density of (18F-RR) VIP was 4-fold greater in the tumor than in the normal tissue with the highest uptake (intestinal) and was ∼15-fold greater than in the normal breast, indicating that (18F-RR) VIP could localize to breast tumors in vivo. However, a comparison of (18F-RR) VIP and 18F-FDG showed that FDG exhibited a 2- to 3-fold greater tumor accumulation and target-to-non-target ratio relative to (18F-RR) VIP (64). To further improve the stability and biodistribution characteristics of 18F-labeled VIP, Cheng et al (65,66) synthesized (R8,15,21, L17) VIP by replacing Asp8, Lys15, and Lys21 with Arg and Met17 with Leu in the amino acid sequence of VIP. The radiolabeled 18F-(R8,15,21, L17) VIP was obtained with high radio-chemical purity, high specific radioactivity and good stability in vitro. Biodistribution data showed higher tumor-to-muscle and tumor-to-blood ratios, indicating its potential application as a PET imaging agent for tumors overexpressing VIPRs.

The radionuclide 64Cu has a longer half-life and a wealth of known chemistry and provides nearly quantitative yields so that the radiolabeled compound can be prepared without further purification (Table I). 64Cu-1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid (TETA) VIP was the first reported 64Cu-VIP analog, but no further preclinical or clinical data have been reported (67). Subsequently, Thakur et al (68) synthesized the VIP analog TP3982 to harbor a carboxy-terminal lysine residue separated from asparagine by 4-aminobutyric acid (Aba) as a spacer. The biological activity of 64Cu-TP3982 was not compromised. It also exhibited in vivo stability and a 74-fold increase in tumor uptake relative to 99mTc-TP3654, which had previously been successfully used to image human tumors. Thus, 64Cu-TP3982 is a desirable PET imaging agent for the imaging of cancers overexpressing VIPRs. Zhang et al (69,70) used the same method to synthesize the VIP analog TP3939 and label it with 64Cu. The imaging data obtained for 64Cu-TP3939 in experimental and spontaneous prostate cancer indicated that with its uncompromised biological activity, 64Cu-TP3939 could detect xenografts and cases of occult prostate cancer that were not detectable with 18F-FDG or CT. Thus, 64Cu-TP3939 is worthy of further investigation for the PET imaging of human tumors and their metastases or recurrent lesions and for determining the efficacy of tumor therapy.

Targeting VIP receptor for cancer molecular therapy

VIP antagonist-based cancer therapy

Since VIP is thought to have growth promoting properties through VIP receptor activation (71,72), the treatment of human tumors with a VIP antagonist may lead to the inhibition of tumor growth (Table II). The VIP-receptor antagonist VIPhyb was synthesized as a hybrid peptide of neurotensin and VIP consisting of an N-terminal Lys-Pro-Arg-Arg-Pro-Tyr (designed to increase membrane permeability), followed by the C-terminal 22 amino acids of VIP (73). This broad spectrum VIP receptor antagonist inhibited non-small cell lung cancer (73), breast cancer (74), and colon cancer (75) growth in vitro and in vivo. In addition, a further modification of VIPhyb was the addition of a stearyl group at the N-terminus and the exchange of the methionine at position 17 with norleucine. These modifications resulted in an antagonist with improved affinity (SN)VIPhyb (76). (SN)VIPhyb bound the VPAC1 receptor with an ∼10-fold greater affinity than VIPhyb and acted as a cytostatic agent in non-small cell lung cancer (77) and pancreatic cancer (Table II) (78). Moreover, (SN)VIPhyb enhanced the anti-proliferative effects of chemotherapeutic agents on cancer cell lines (79).

Table II.

Some analogs of VIP and their therapeutic effects.

Table II.

Some analogs of VIP and their therapeutic effects.

AnalogsSelectivityAffinity (IC50:nM)Application
VIPhybPAC antagonist300Inhibition of colon and breast cancer
(SN)VIPhybVPAC1 antagonist30Inhibition of non-small lung cancer
[K15, R16, L27]-VIP(1–7)/GRF (8–27)VPAC1 antagonist10Inhibition various cancers
VIP-LALA-EVPAC1 agonist100Inhibition of breast cancer
Ro 25-1553VPAC2 agonist1Therapeutic for bronchial athma
BAY 55-9837VPAC2 agonist60Stimulation of insulin secretion
stearyl-[Nle17]-VIPNonselective agonist5Inhibition of breast cancer
[R15, 20, 21, L17]-VIP-GRRNonselective agonist1.4Treatment of asthma and COPD
[R15, 20, 21, L17]-VIP(1–23)Nonselective agonist48Relaxation of muscle

To avoid the side effects of the broad-spectrum VIP receptor antagonists, new analogs selectively inhibiting each VIP receptor subtype may significantly prevent undesirable side effects, such as the inhibition of VPAC2 activation, which has been associated with glucose-dependent insulin secretion and bronchodilation (80). Recently, a VPAC1 antagonist conjugated to a high-molecular-weight PEG was synthesized, and the pharmacokinetic characteristics were improved without compromising functional activities. The results showed that the selective inhibition of the VPAC1 receptor is sufficient to prevent tumor proliferation, which suggests that a VPAC1-selective antagonist may be a safe and effective tool for treating tumors (unpublished).

Cytotoxic peptide conjugate-based cancer therapy

Conventional chemotherapy is limited by multidrug resistance of tumor cells, toxicity to normal cells and a lack of tumor specificity. The more specific delivery of chemotherapeutic drugs to cancer cells can produce a higher drug concentration in tumors and reduce the toxicity to normal cells (81). Therefore, ‘targeted therapy’ was introduced with the aim of enhancing the specificity of chemotherapy and reducing the non-specific toxicity to normal cells (82). Since high-affinity VIP receptors are overexpressed on many tumors, they can be targeted using cytotoxic VIP conjugates.

A new cytotoxic analog of VIP-ellipticine was prepared that consisted of tetra- and pentapeptide ellipticine (E) attached to the C-terminus of VIP. The VIP-E conjugates functioned as VPAC1 receptor agonists, bound to VPAC1 receptors with high affinity and retained their anti-proliferative activity. The VIP-E conjugates were internalized by cancer cells expressing the VPAC1 receptor and were subsequently metabolized by proteolytic enzymes, leading to the release of ellipticine and cellular cytotoxicity. The VIP-E derivatives exhibited significant cytotoxicity toward breast cancer cells and lung cancer cells in vitro (83,84). Subsequently, Moody et al (85) extended these studies and synthesized VIP-camptothecin (CPT) conjugates that contain a novel carbamate linker with a built-in nucleophile associated releasing group, L2. The obtained conjugate (A-NL-K)-VIP-L2-CPT is metabolized by cytochrome P450 enzymes, releases CPT and exhibits cytotoxic effects on breast cancer cells.

Peptide-receptor radionuclide therapy (PRRT)

As mentioned previously, VIP receptor-positive tumors can be targeted with radiolabeled VIP and its analogs (52,61). It is theoretically possible to treat such tumors selectively with therapeutic nuclide-labeled VIP analogs. This peptide-receptor radio-nuclide therapy is based on the presence of high levels of VIP receptors in tumors and on their ability to form ligand-receptor complexes, which allows for the internalization and accumulation of the radiopharmaceuticals inside the tumors. Although there are many studies regarding somatostatin receptor-based radiotherapy (51,86), there are currently no reports on VIP receptor radiotherapy for human tumors. One reason may be the lack of appropriate radiolabeled ligands. A second reason is certainly the inadequate tumor-to-non-tumor ratio for radiotherapy. In this case, VIP receptor-based PRRT could be highly radiotoxic to surrounding normal tissues, especially to radiosensitive tissues such as immune, lung, kidney and liver cells (51). Therefore, the development of new peptide analogs with increased binding affinity and specificity may lead to a higher accumulation of radioactivity in tumors and improve the efficacy of peptide receptor radionuclide therapy.

Prospective and Conclusion

VIP receptors are very important in the biology of many malignancies and are overexpressed in many tumors; thus, VIP receptors can be targeted by receptor-specific molecules. The generation of radiolabeled peptides has opened new avenues for the molecular imaging and therapy of tumors. Due to their low molecular weight, high-affinity, and good tissue/cell penetration, they are becoming ideal candidates for molecular imaging techniques and therapeutic interventions. To date, radiolabeled peptides specific for VIP receptors have evolved from the initial native VIP peptide into peptide analogs or peptide antagonists with improved pharmacokinetic profiles. Although a series of preclinical studies on VIP receptor-based imaging and therapy using radionuclide-labeled VIP and its analogs, antagonists and cytotoxic peptide conjugates have shown promising results, the therapeutic potential of VIP receptor-based radionuclide therapy must be refined and optimized.

Under the circumstances, it is necessary to develop novel peptide analogs with improved binding affinity, specificity and stability to result in a higher accumulation of radioactivity inside tumor cells. In a recent study, our research group identified a novel dodecapeptide that could specifically bind to the VPAC1 receptor with high affinity using a phage display peptide library. The results imply that the peptide may serve as a potential molecular imaging probe and therapeutic agent (87). Increasing the therapeutic window, which can be achieved by reducing the radiation toxicity to normal organs, could significantly enhance the therapeutic effects through increased injected radioactivity. Since the kidney has been one of the dose-limiting organs in some clinical studies, several standard procedures to reduce renal uptake have been developed and followed (88). Most importantly, the combination of radionuclide imaging and therapy with other diagnosis and treatment modalities, such as CT, MRI and chemotherapy, may greatly increase the efficiency of early diagnosis and treatment for the heterogeneous tumors.

Recently, the most common somatostatin receptor-based agent 111In-DTPA-pentetreotide (Octreoscan; Mallinckrodt) was approved by the US Food and Drug Administration for clinical somatostatin receptor imaging (89). Moreover, several 90Y-labeled octreotides were tested in different phase-I and phase-II clinical trials (90), and a major breakthrough in the tumor receptor-targeted imaging and therapy field may be on the way. The clinical success of somatostatin receptor imaging and therapy will provide insight into the exploration and development of VIP receptor-based imaging and therapy. Although there are still many other drawbacks to overcome in VIP receptor-based imaging and therapy, we believe that major progress will be made in preclinical settings. The importance of VIP receptors in tumor biology and the ability to predict responses to targeted therapy and monitor drug interventions suggest that VIP receptor imaging will be critical for oncologic molecular imaging and will play a key role in cancer management in the future.

Abbreviations:

VIP

vasoactive intestinal peptide;

PACAP

pituitary adenylate cyclase-activating polypeptide;

PHM

peptide histidine methionine amide;

GRF

growth-hormone-releasing-factor;

PHI

peptide histidine isoleucine;

GPCR

G-protein-coupled receptor;

SPECT

single photon emission computed tomography;

PET

positron emission tomography;

CT

computed tomography;

MRI

magnetic resonance imaging;

FDG

fluorodeoxyglucose;

PRRT

peptide-receptor radionuclide therapy

References

1. 

Ozben T: Mechanisms and strategies to overcome multiple drug resistance in cancer. FEBS Lett. 580:2903–2909. 2006. View Article : Google Scholar : PubMed/NCBI

2. 

Xie X, Tang B, Zhou J, Gao Q and Zhang P: Inhibition of the PI3K/Akt pathway increases the chemosensitivity of gastric cancer to vincristine. Oncol Rep. 30:773–782. 2013.PubMed/NCBI

3. 

Pluchino KM, Hall MD, Goldsborough AS, Callaghan R and Gottesman MM: Collateral sensitivity as a strategy against cancer multidrug resistance. Drug Resist Updat. 15:98–105. 2012. View Article : Google Scholar : PubMed/NCBI

4. 

Burris HA III: Overcoming acquired resistance to anticancer therapy: focus on the PI3K/AKT/mTOR pathway. Cancer Chemother Pharmacol. 71:829–842. 2013. View Article : Google Scholar : PubMed/NCBI

5. 

Garanger E, Boturyn D and Dumy P: Tumor targeting with RGD peptide ligands-design of new molecular conjugates for imaging and therapy of cancers. Anticancer Agents Med Chem. 7:552–558. 2007. View Article : Google Scholar : PubMed/NCBI

6. 

Tolmachev V: Imaging of HER-2 overexpression in tumors for guiding therapy. Curr Pharm Des. 14:2999–3019. 2008. View Article : Google Scholar : PubMed/NCBI

7. 

Kaklamani V and O’Regan RM: New targeted therapies in breast cancer. Semin Oncol. 31:20–25. 2004. View Article : Google Scholar

8. 

Mankoff DA, Link JM, Linden HM, Sundararajan L and Krohn KA: Tumor receptor imaging. J Nucl Med. 49:S149–S163. 2008. View Article : Google Scholar

9. 

Nunn AD: Molecular imaging and personalized medicine: an uncertain future. Cancer Biother Radiopharm. 22:722–739. 2007. View Article : Google Scholar : PubMed/NCBI

10. 

Mankoff DA: A definition of molecular imaging. J Nucl Med. 48:N18–N21. 2007.

11. 

Sherwood NM, Krueckl SL and McRory JE: The origin and function of the pituitary adenylate cyclase-activating polypeptide (PACAP)/glucagon superfamily. Endocr Rev. 21:619–670. 2000.PubMed/NCBI

12. 

Said SI and Mutt V: Polypeptide with broad biological activity: isolation from small intestine. Science. 169:1217–1218. 1970. View Article : Google Scholar : PubMed/NCBI

13. 

Piper PJ, Said SI and Vane JR: Effects on smooth muscle preparations of unidentified vasoactiv peptides from intestine and lung. Nature. 225:1144–1146. 1970. View Article : Google Scholar : PubMed/NCBI

14. 

Barbezat GO and Grossman MI: Intestinal secretion: stimulation by peptides. Science. 174:422–424. 1971. View Article : Google Scholar : PubMed/NCBI

15. 

Gozes I, Fridkinb M, Hill JM and Brenneman DE: Pharmaceutical VIP: prospects and problems. Curr Med Chem. 6:1019–1034. 1999.PubMed/NCBI

16. 

Gozes I and Furman S: VIP and drug design. Curr Pharm Des. 9:483–494. 2003. View Article : Google Scholar : PubMed/NCBI

17. 

Tsukada T, Horovitch SJ, Montminy MR, Mandel G and Goodman RH: Structure of the human vasoactive intestinal polypeptide gene. DNA. 4:293–300. 1985.

18. 

Gozes I, Avidor R, Yahav Y, Katznelson D, Croce CM and Huebner K: The gene encoding vasoactive intestinal peptide is located on human chromosome 6p21-6qter. Hum Genet. 75:41–44. 1987.PubMed/NCBI

19. 

Davidson A, Moody TW and Gozes I: Regulation of VIP gene expression in general. Human lung cancer cells in particular. J Mol Neurosci. 7:99–110. 1996. View Article : Google Scholar : PubMed/NCBI

20. 

Itoh N, Obata K, Yanaihara N and Okamoto H: Human preprovasoactive intestinal polypeptide contains a novel PHI-27-like peptide, PHM-27. Nature. 304:547–549. 1983. View Article : Google Scholar : PubMed/NCBI

21. 

Bodner M, Fridkin M and Gozes I: Coding sequences for vasoactive intestinal peptide and PHM-27 peptide are located on two adjacent exons in the human genome. Proc Natl Acad Sci USA. 82:3548–3551. 1985. View Article : Google Scholar : PubMed/NCBI

22. 

Vandermeers A, Vandenborre S, Hou X, de Neef P, Robberecht P, Vandermeers-Piret MC and Christophe J: Antagonistic properties are shifted back to agonistic properties by further N-terminal shortening of pituitary adenylate-cyclase-activating peptides in human neuroblastoma NB-OK-1 cell membranes. Eur J Biochem. 208:815–819. 1992. View Article : Google Scholar

23. 

Vaudry D, Gonzalez BJ, Basille M, Yon L, Fournier A and Vaudry H: Pituitary adenylate cyclase-activating polypeptide and its receptors: from structure to functions. Pharmacol Rev. 52:269–324. 2000.PubMed/NCBI

24. 

Fahrenkrug J: VIP and PACAP. Results Probl Cell Differ. 50:221–234. 2010.

25. 

Dickson L and Finlayson K: VPAC and PAC receptors: From ligands to function. Pharmacol Ther. 121:294–316. 2009. View Article : Google Scholar : PubMed/NCBI

26. 

Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BK, Hashimoto H, Galas L and Vaudry H: Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev. 61:283–357. 2009.PubMed/NCBI

27. 

Muller JM, Debaigt C, Goursaud S, Montoni A, Pineau N, Meunier AC and Janet T: Unconventional binding sites and receptors for VIP and related peptides PACAP and PHI/PHM: an update. Peptides. 28:1655–1666. 2007. View Article : Google Scholar : PubMed/NCBI

28. 

Laburthe M, Couvineau A and Marie JC: VPAC receptors for VIP and PACAP. Recept Chann. 8:137–153. 2002. View Article : Google Scholar : PubMed/NCBI

29. 

Couvineau A, Lacapere JJ, Tan YV, Rouyer-Fessard C, Nicole P and Laburthe M: Identification of cytoplasmic domains of hVPAC1 receptor required for activation of adenylyl cyclase. Crucial role of two charged amino acids strictly conserved in class II G protein-coupled receptors. J Biol Chem. 278:24759–24766. 2003. View Article : Google Scholar

30. 

Dickson L, Aramori I, McCulloch J, Sharkey J and Finlayson K: A systematic comparison of intracellular cyclic AMP and calcium signalling highlights complexities in human VPAC/PAC receptor pharmacology. Neuropharmacology. 51:1086–1098. 2006. View Article : Google Scholar

31. 

Barrie AP, Clohessy AM, Buensuceso CS, Rogers MV and Allen JM: Pituitary adenylyl cyclase-activating peptide stimulates extracellular signal-regulated kinase 1 or 2 (ERK1/2) activity in a Ras-independent, mitogen-activated protein Kinase/ERK kinase 1 or 2-dependent manner in PC12 cells. J Biol Chem. 272:19666–19671. 1997. View Article : Google Scholar

32. 

Lelièvre V, Pineau N, Du J, Wen CH, Nguyen T, Janet T, Muller JM and Waschek JA: Differential effects of peptide histidine isoleucine (PHI) and related peptides on stimulation and suppression of neuroblastoma cell proliferation. A novel VIP-independent action of PHI via MAP kinase. J Biol Chem. 273:19685–19690. 1998.PubMed/NCBI

33. 

Delgado M and Ganea D: Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide inhibit interleukin-12 transcription by regulating nuclear factor kappaB and Ets activation. J Biol Chem. 274:31930–31940. 1999. View Article : Google Scholar

34. 

Hashimoto H, Shintani N, Tanaka K, Mori W, Hirose M, Matsuda T, Sakaue M, Miyazaki J, Niwa H, Tashiro F, Yamamoto K, Koga K, Tomimoto S, Kunugi A, Suetake S and Baba A: Altered psychomotor behaviors in mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP). Proc Natl Acad Sci USA. 98:13355–13360. 2001. View Article : Google Scholar : PubMed/NCBI

35. 

Jozsa R, Hollosy T, Nemeth J, Tamás A, Lubics A, Jakab B, Olah A, Arimura A and Reglödi D: Presence of PACAP and VIP in embryonic chicken brain. Ann NY Acad Sci. 1070:348–353. 2006. View Article : Google Scholar : PubMed/NCBI

36. 

Valdehita A, Carmena MJ, Collado B, Prieto JC and Bajo AM: Vasoactive intestinal peptide (VIP) increases vascular endothelial growth factor (VEGF) expression and secretion in human breast cancer cells. Regul Pept. 144:101–108. 2007. View Article : Google Scholar : PubMed/NCBI

37. 

Valdehita A, Bajo AM, Schally AV, Varga JL, Carmena MJ and Prieto JC: Vasoactive intestinal peptide (VIP) induces transactivation of EGFR and HER2 in human breast cancer cells. Mol Cell Endocrinol. 302:41–48. 2009. View Article : Google Scholar : PubMed/NCBI

38. 

Sreedharan SP, Patel DR, Huang JX and Goetzl EJ: Cloning and functional expression of a human neuroendocrine vasoactive intestinal peptide receptor. Biochem Biophys Res Commun. 193:546–553. 1993. View Article : Google Scholar : PubMed/NCBI

39. 

Usdin TB, Bonner TI and Mezey E: Two receptors for vasoactive intestinal polypeptide with similar specificity and complementary distributions. Endocrinology. 135:2662–2680. 1994.PubMed/NCBI

40. 

Wei Y and Mojsov S: Tissue specific expression of different human receptor types for pituitary adenylate cyclase activating polypeptide and vasoactive intestinal polypeptide: implications for their role in human physiology. J Neuroendocrinol. 8:811–817. 1996. View Article : Google Scholar

41. 

Moller K and Sundler F: Expression of pituitary adenylate cyclase activating peptide (PACAP) and PACAP type I receptors in the rat adrenal medulla. Regul Pept. 63:129–139. 1996. View Article : Google Scholar : PubMed/NCBI

42. 

Zeng N, Kang T, Lyu RM, Wong H, Wen Y, Walsh JH, Sachs G and Pisegna JR: The pituitary adenylate cyclase activating polypeptide type 1 receptor (PAC1-R) is expressed on gastric ECL cells: evidence by immunocytochemistry and RT-PCR. Ann NY Acad Sci. 865:147–156. 1998. View Article : Google Scholar : PubMed/NCBI

43. 

Reubi JC, Läderach U, Waser B, Gebbers JO, Robberecht P and Laissue JA: Vasoactive intestinal peptide/pituitary adenylate cyclase-activating peptide receptor subtypes in human tumors and their tissues of origin. Cancer Res. 60:3105–3112. 2000.

44. 

Reubi JC, Körner M, Waser B, Mazzucchelli L and Guillou L: High expression of peptide receptors as a novel target in gastrointestinal stromal tumours. Eur J Nucl Med Mol Imaging. 31:803–810. 2004. View Article : Google Scholar : PubMed/NCBI

45. 

Gozes I and Furman S: Clinical endocrinology and metabolism. Potential clinical applications of vasoactive intestinal peptide: a selected update. Best Pract Res Clin Endocrinol Metab. 18:623–640. 2004. View Article : Google Scholar : PubMed/NCBI

46. 

Whitmarsh AJ and Davis RJ: Transcription factor AP-1 regulation by mitogen-activated protein kinase signal transduction pathways. J Mol Med. 74:589–607. 1996. View Article : Google Scholar : PubMed/NCBI

47. 

Casibang M, Purdom S, Jakowlew S, Neckers L, Zia F, Ben-Av P, Hla T, You L, Jablons DM and Moody TW: Prostaglandin E2 and vasoactive intestinal peptide increase vascular endothelial cell growth factor mRNAs in lung cancer cells. Lung Cancer. 31:203–212. 2001.

48. 

Mankoff DA, O’Sullivan F, Barlow WE and Krohn KA: Molecular imaging research in the outcomes era: measuring outcomes for individualized cancer therapy. Acad Radiol. 14:398–405. 2007. View Article : Google Scholar : PubMed/NCBI

49. 

Goldenberg DM, DeLand F, Kim E, Bennett S, Primus FJ, van Nagell JR Jr, Estes N, DeSimone P and Rayburn P: Use of radiolabeled antibodies to carcinoembryonic antigen for the detection and localization of diverse cancers by external photo-scanning. N Engl J Med. 298:1384–1386. 1978. View Article : Google Scholar : PubMed/NCBI

50. 

Behr TM, Memtsoudis S, Sharkey RM, Blumenthal RD, Dunn RM, Gratz S, Wieland E, Nebendahl K, Schmidberger H, Goldenberg DM and Becker W: Experimental studies on the role of antibody fragments in cancer radio-immunotherapy: influence of radiation dose and dose rate on toxicity and anti-tumor efficacy. Int J Cancer. 77:787–795. 1998. View Article : Google Scholar : PubMed/NCBI

51. 

Reubi JC: Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocr Rev. 24:389–427. 2003. View Article : Google Scholar : PubMed/NCBI

52. 

Virgolini I, Raderer M, Kurtaran A, Angelberger P, Banyai S, Yang Q, Li S, Banyai M, Pidlich J, Niederle B, Scheithauer W and Valent P: Vasoactive intestinal peptide-receptor imaging for the localization of intestinal adenocarcinomas and endocrine tumors. N Engl J Med. 33:1116–1121. 1994. View Article : Google Scholar : PubMed/NCBI

53. 

Raderer M, Kurtaran A, Hejna M, Vorbeck F, Angelberger P, Scheithauer W and Virgolini I: 123I-labelled vasoactive intestinal peptide receptor scintigraphy in patients with colorectal cancer. Br J Cancer. 78:1–5. 1998. View Article : Google Scholar

54. 

Raderer M, Kurtaran A, Yang Q, Meghdadi S, Vorbeck F, Hejna M, Angelberger P, Kornek G, Pidlich J, Scheithauer W and Virgolini I: Iodine-123-vasoactive intestinal peptide receptor scanning in patients with pancreatic cancer. J Nucl Med. 39:1570–1575. 1998.PubMed/NCBI

55. 

Virgolini I, Kurtaran A, Leimer M, Kaserer K, Peck-Radosavljevic M, Angelberger P, Hübsch P, Dvorak M, Valent P and Niederle B: Location of a VIPoma by iodine-123-vasoactive intestinal peptide scintigraphy. J Nucl Med. 39:1575–1579. 1998.PubMed/NCBI

56. 

Virgolini I, Kurtaran A, Raderer M, Leimer M, Angelberger P, Havlik E, Li S, Scheithauer W, Niederle B and Valent P: Vasoactive intestinal peptide receptor scintigraphy. J Nucl Med. 36:1732–1739. 1995.PubMed/NCBI

57. 

Raderer M, Becherer A, Kurtaran A, Angelberger P, Li S, Leimer M, Weinlaender G, Kornek G, Kletter K, Scheithauer W and Virgolini I: Comparison of iodine-123-vasoactive intestinal peptide receptor scintigraphy and indium-111-CYT-103 immunoscintigraphy. J Nucl Med. 37:1480–1487. 1996.PubMed/NCBI

58. 

Thakur ML, Marcus CS, Saeed S, Pallela V, Minami C, Diggles L, Le Pham H, Ahdoot R and Kalinowski EA: 99mTc-labeled vasoactive intestinal peptide analog for rapid localization of tumors in humans. J Nucl Med. 41:107–110. 2000.

59. 

Pallela VR, Thakur ML, Chakder S and Rattan S: 99mTc-labeled vasoactive intestinal peptide receptor agonist: functional studies. J Nucl Med. 40:352–360. 1999.

60. 

Thakur ML, Marcus CS, Saeed S, Pallela V, Minami C, Diggles L, Pham HL, Ahdoot R, Kalinowski EA and Moody T: Imaging tumors in humans with Tc-99m-VIP. Ann NY Acad Sci. 921:37–44. 2000. View Article : Google Scholar : PubMed/NCBI

61. 

Rao PS, Thakur ML, Pallela V, Patti R, Reddy K, Li H, Sharma S, Pham HL, Diggles L, Minami C and Marcus CS: 99mTc labeled VIP analog: evaluation for imaging colorectal cancer. Nucl Med Biol. 28:445–450. 2001. View Article : Google Scholar

62. 

Kothari K, Prasad S, Korde A, Mukherjee A, Mathur A, Jaggi M, Venkatesh M, Pillai AM, Mukherjee R and Ramamoorthy N: 99mTc(CO)3-VIP analogues: preparation and evaluation as tumor imaging agent. Appl Radiat Isot. 65:382–386. 2007. View Article : Google Scholar

63. 

Moody TW, Leyton J, Unsworth E, John C, Lang L and Eckelman WC: (Arg15, Arg21) VIP: evaluation of biological activity and localization to breast cancer tumors. Peptides. 19:585–592. 1998.

64. 

Jagoda EM, Aloj L, Seidel J, Lang L, Moody TW, Green S, Caraco C, Daube-Witherspoon M, Green MV and Eckelman WC: Comparison of an 18F labeled derivative of vasoactive intestinal peptide and 2-deoxy-2-[18F]fluoro-D-glucose in nude mice bearing breast cancer xenografts. Mol Imaging Biol. 4:369–379. 2002.

65. 

Cheng D, Yin D, Zhang L, Wang M, Li G and Wang Y: Radiosynthesis of 18F-(R8,15,21, L17)-vasoactive intestinal peptide and preliminary evaluation in mice bearing C26 colorectal tumours. Nucl Med Commun. 28:501–506. 2007.

66. 

Cheng D, Yin D, Li G, Wang M, Li S, Zheng M, Cai H and Wang Y: Radiolabeling and in vitro and in vivo characterization of [18F]FB-[R8,15,21, L17]-VIP as a PET imaging agent for tumor overexpressed VIP receptors. Chem Biol Drug Des. 68:319–325. 2006.

67. 

Chen X, Edwards WB, Anderson CJ, Mccarthy TJ and Welch MJ: Solid phase synthesis of TETA conjugated vasoactive intestinal peptide and in vivo behavior of copper-64 radiolabeled VIP conjugate. J Labelled Compds Radiopharm. 44:S688–S690. 2001. View Article : Google Scholar

68. 

Thakur ML, Aruva MR, Gariepy J, Acton P, Rattan S, Prasad S, Wickstrom E and Alavi A: PET imaging of oncogene over-expression using 64Cu-vasoactive intestinal peptide (VIP) analog: comparison with 99mTc-VIP analog. J Nucl Med. 45:1381–1389. 2004.PubMed/NCBI

69. 

Zhang K, Aruva MR, Shanthly N, Cardi CA, Rattan S, Patel C, Kim C, McCue PA, Wickstrom E and Thakur ML: PET imaging of VPAC1 expression in experimental and spontaneous prostate cancer. J Nucl Med. 49:112–121. 2008. View Article : Google Scholar : PubMed/NCBI

70. 

Zhang K, Aruva MR, Shanthly N, Cardi CA, Patel CA, Rattan S, Cesarone G, Wickstrom E and Thakur ML: Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating peptide (PACAP) receptor specific peptide analogues for PET imaging of breast cancer: In vitro/in vivo evaluation. Regul Pept. 144:91–100. 2007. View Article : Google Scholar

71. 

Collado B, Carmena MJ, Clemente C, Prieto JC and Bajo AM: Vasoactive intestinal peptide enhances growth and angiogenesis of human experimental prostate cancer in a xenograft model. Peptides. 28:1896–1901. 2007. View Article : Google Scholar : PubMed/NCBI

72. 

Fernández-Martínez AB, Bajo AM, Sánchez-Chapado M, Prieto JC and Carmena MJ: Vasoactive intestinal peptide behaves as a pro-metastatic factor in human prostate cancer cells. Prostate. 69:774–786. 2009.PubMed/NCBI

73. 

Moody TW, Zia F, Draoui M, Brenneman DE, Fridkin M, Davidson A and Gozes I: A vasoactive intestinal peptide antagonist inhibits non-small cell lung cancer growth. Proc Natl Acad Sci USA. 90:4345–4349. 1993. View Article : Google Scholar : PubMed/NCBI

74. 

Zia H, Hida T, Jakowlew S, Birrer M, Gozes Y, Reubi JC, Fridkin M, Gozes I and Moody TW: Breast cancer growth is inhibited by vasoactive intestinal peptide (VIP) hybrid, a synthetic VIP receptor antagonist. Cancer Res. 56:3486–3489. 1996.PubMed/NCBI

75. 

Levy A, Gal R, Granoth R, Dreznik Z, Fridkin M and Gozes I: In vitro and in vivo treatment of colon cancer by VIP antagonists. Regul Pept. 109:127–133. 2002. View Article : Google Scholar : PubMed/NCBI

76. 

Moody TW, Jensen RT, Fridkin M and Gozes I: (N-stearyl, norleucine 17) VIP hybrid is a broad spectrum vasoactive intestinal peptide receptor antagonist. J Mol Neurosci. 18:29–35. 2002. View Article : Google Scholar : PubMed/NCBI

77. 

Moody TW, Leyton J, Coelho T, Jakowlew S, Takahashi K, Jameison F, Koh M, Fridkin M, Gozes I and Knight M: (Stearyl, Norleucine 17) VIP hybrid antagonizes VIP receptors on non-small cell lung cancer cells. Life Sci. 61:1657–1666. 1997. View Article : Google Scholar : PubMed/NCBI

78. 

Zia H, Leyton J, Casibang M, Hau V, Brenneman D, Fridkin M, Gozes I and Moody TW: (N-stearyl, norleucine17) VIP hybrid inhibits the growth of pancreatic cancer cell lines. Life Sci. 66:379–387. 2000. View Article : Google Scholar : PubMed/NCBI

79. 

Moody TW, Leyton J, Chan D, Brenneman DC, Fridkin M, Gelber E, Levy A and Gozes I: VIP receptor antagonists and chemotherapeutic drugs inhibit the growth of breast cancer cells. Breast Cancer Res Treat. 68:55–64. 2001. View Article : Google Scholar : PubMed/NCBI

80. 

Pan CQ, Hamren S, Roczniak S, Tom I and DeRome M: Generation of PEGylated VPAC1-selective antagonists that inhibit proliferation of a lung cancer cell line. Peptides. 29:479–486. 2008. View Article : Google Scholar : PubMed/NCBI

81. 

Schally AV and Nagy A: Chemotherapy targeted to cancers through tumoral hormone receptors. Trends Endocrinol Metab. 15:300–310. 2004. View Article : Google Scholar : PubMed/NCBI

82. 

Kim JA: Targeted therapies for the treatment of cancer. Am J Surg. 186:264–268. 2003. View Article : Google Scholar : PubMed/NCBI

83. 

Moody TW, Czerwinski G, Tarasova NI and Michejda CJ: VIP-ellipticine derivatives inhibit the growth of breast cancer cells. Life Sci. 71:1005–1014. 2002. View Article : Google Scholar : PubMed/NCBI

84. 

Moody TW, Czerwinski G, Tarasova NI, Moody DL and Michejda CJ: The development of VIP-ellipticine conjugates. Regul Pept. 123:187–192. 2004. View Article : Google Scholar : PubMed/NCBI

85. 

Moody TW, Mantey SA, Fuselier JA, Coy DH and Jensen RT: Vasoactive intestinal peptide-camptothecin conjugates inhibit the proliferation of breast cancer cells. Peptides. 28:1883–1890. 2007. View Article : Google Scholar : PubMed/NCBI

86. 

Zaccaro L, Del Gatto A, Pedone C and Saviano M: Peptides for tumour therapy and diagnosis: current status and future directions. Curr Med Chem. 16:780–795. 2009. View Article : Google Scholar : PubMed/NCBI

87. 

Tang B, Li Z, Huang D, Zheng L and Li Q: Screening of a specific peptide binding to VPAC1 receptor from a phage display peptide library. PLoS One. 8:e542642013. View Article : Google Scholar : PubMed/NCBI

88. 

de Visser M, Verwijnen SM and de Jong M: Update: improvement strategies for peptide receptor scintigraphy and radionuclide therapy. Cancer Biother Radiopharm. 23:137–157. 2008.PubMed/NCBI

89. 

Balon HR, Goldsmith SJ, Siegel BA, Silberstein EB, Krenning EP, Lang O and Donohoe KJ: Procedure guideline for somatostatin receptor scintigraphy with (111)In-pentetreotide. J Nucl Med. 42:1134–1138. 2001.PubMed/NCBI

90. 

Kwekkeboom DJ, Kam BL, van Essen M, Teunissen JJ, van Eijck CH, Valkema R, de Jong M, de Herder WW and Krenning EP: Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer. 17:R53–R73. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

2014-April
Volume 44 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tang B, Yong X, Xie R, Li Q and Yang S: Vasoactive intestinal peptide receptor-based imaging and treatment of tumors (Review). Int J Oncol 44: 1023-1031, 2014
APA
Tang, B., Yong, X., Xie, R., Li, Q., & Yang, S. (2014). Vasoactive intestinal peptide receptor-based imaging and treatment of tumors (Review). International Journal of Oncology, 44, 1023-1031. https://doi.org/10.3892/ijo.2014.2276
MLA
Tang, B., Yong, X., Xie, R., Li, Q., Yang, S."Vasoactive intestinal peptide receptor-based imaging and treatment of tumors (Review)". International Journal of Oncology 44.4 (2014): 1023-1031.
Chicago
Tang, B., Yong, X., Xie, R., Li, Q., Yang, S."Vasoactive intestinal peptide receptor-based imaging and treatment of tumors (Review)". International Journal of Oncology 44, no. 4 (2014): 1023-1031. https://doi.org/10.3892/ijo.2014.2276