CHAPTER 16
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer Ryan Clay, Shadab A. Siddiqi Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
Abstract Pancreatic adenocarcinoma is one of the most aggressive cancers and has a rising incidence and high mortality. The high mortality rate is attributed to a lack of adequate diagnostics, which limits the efficacy of the few currently available treatment options. The poor prognosis of pancreatic cancer emphasizes an increasing demand to develop efficient diagnostic markers and therapeutic approaches. The current review focuses on advances made in the development of robust molecular biomarkers that can be used for diagnosis and targeted therapies. Here, we discuss three major types of molecular markers for the detection of pancreatic adenocarcinoma: serum, tumor tissue, and cancer stem cell markers. Substantial advances have been made to improve our understanding of pancreatic adenocarcinoma at both the cellular and molecular levels, strengthening the possibilities of early detection and targeted therapies; they are discussed in this chapter.
Keywords: Intraductal papillary mucinous neoplasms (IPMNs); Mucinous cystic neoplasms (MCNs); Pancreatic cancer (PC); Pancreatic ductal adenocarcinoma (PDAC); Pancreatic intraepithelial neoplasms (PanINs).
Background Despite an incidence of only 3.2%, pancreatic cancer (PC) is third in overall cancer deaths with an average 5-year survival of only 8.5% [1]. However, mortality can vary widely depending on the size and stage of the tumor at the time of detection [2,3], with 5-year survival of over 80% for cancers smaller than 10 mm [3]. Despite the advantage of early detection for prognosis and survival, the lack of molecular markers for detecting early or premetastatic PC combined with the invasiveness and cost of existing screening methods prohibit routine screening [2e7]. As a result, over 80% of cancers have already metastasized upon initial discovery [1,3]. G.P. Nagaraju, S. Ahmad (eds.) Theranostic Approach for Pancreatic Cancer ISBN 978-0-12-819457-7 https://doi.org/10.1016/B978-0-12-819457-7.00016-5
Copyright © 2019 Elsevier Inc. All rights reserved.
325
326
Theranostic Approach for Pancreatic Cancer
More advanced molecular diagnostics and treatments are necessary to reduce the burden of this disease. Exocrine tumors of the pancreas are by far the most common (over 95%) [8], with pancreatic ductal adenocarcinoma (PDAC) making up over 90% of those cases [9]. Other, rarer types of PC include acinar cell carcinomas, pancreatoblastomas, and neuroendocrine tumors [10,11]. Despite improvements in diagnostics and therapeutics in other cancers, the process of detecting PC remains difficult and relies largely on visual screening methods [4,5,7]. Although the average exocrine tumor will take more than a decade to metastasize [12], both early-stage cancers and the precancerous lesions from which they arise are largely asymptomatic and, in the case of pancreatic intraepithelial neoplasms (PanINs), they can be too small to detect reliably with magnetic resonance imaging (MRI) or ultrasound [3,5,13]. Most cases are discovered after the manifestation of nonspecific symptoms associated with late-stage metastatic tumors, such as jaundice, abdominal pain, and weight loss [3e5]. Less than 10% of PCs are discovered before they have spread beyond the pancreas, and are often discovered inadvertently or because the tumor has formed in close proximity to and has compressed the common bile duct, pancreatic duct, or mesenteric and celiac nerves, producing symptoms at an earlier stage [14e16].
Pathogenesis of pancreatic ductal adenocarcinoma PDAC is known to arise from one of three precursor lesions: PanINs, intraductal papillary mucinous neoplasms (IPMNs), and mucinous cystic neoplasms (MCNs) [3,13,17]. PanINs are the most common of the three and their progression into PDAC has been well-characterized [3,13,18,19]. They are a promising target for early detection for multiple reasons: they are relatively common and usually benign [3,13,18]; of the estimated 1% of PanINs that will metastasize [18], they will take well over a decade on average to do so [12]; and those that do progress tend to accumulate characteristic mutations and undergo morphological changes in a predictable manner [3,11e13,17,19e21]. There are four hallmark genetic alterations in the progression of PanIN to PDAC: a gain of function mutation in KRAS (84%e96% of PDACs), and the downregulation of tumor suppression genes p16/CDKN2A (90%e95%), TP53 (40%e90%), and SMAD4/DPC4 (50%e55%) [3,7,9,11e13,16,17,22e25]. Interestingly, the pathogenesis of both familial PC, which accounts for less than 10% of cases, and sporadic PC is driven by
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
327
these same four genes [3,19,22]. KRAS mutates early in PanIN pathogenesis and is associated with grade 1 PanINs [3,11,13,17,19]. The most common mutation occurs in codon 12 of exon 2 (with other common mutations in codon 13 and 61), inhibiting KRAS’s GTPase activity and causing it to become constitutively active [11,13,17]. KRAS constitutive activation drives carcinogenesis through the activation of multiple wellknown downstream pathways related to cell growth and proliferation, including the RAFemitogen-activated protein kinase (MAPK) and PI3KAKT signaling cascades [11,17]. It also causes the shortening of telomeres, leading to chromosomal instability [11,13,17]. Next, inactivation of the CDK-inhibiting p16/CDKN2A is seen in PanIN-2 and allows for unchecked progression through the G1-S phase checkpoint [11,13,17]. Later, the loss of TP53 and SMAD4/DPC4 in PanIN-3 results in the loss of DNA damage-induced G2-M cell cycle arrest and the interrupted transduction of proapoptotic signals by transforming growth factor b (TGF-b) and activin, respectively [11,13,17]. Many other less common driver mutations have been reported, including in known protooncogenes Her2 and Akt2 and tumor suppressor genes BRCA1/2, TGFBR1/2, FHIT, serine/threonineprotein kinase 11 (STK11), and ALK4 [16,17,22]. There is variability among the quantity of driver mutations accumulated between tumor cells, with the number of accumulated mutations negatively affecting prognosis [7,26]. IPMNs and MCNs are thought to progress molecularly in a manner similar to that of PanINs, but with some minor differences that will be relevant in the development of future screening and treatment techniques. IPMNs will exhibit loss of function of STK11/liver kinase B1 more commonly than SMAD4, and may express mucin-2 (MUC2) rather than MUC1, which is overexpressed on most PanINs [13,17]. IPMNs also will tend to possess mutations in GNAS either instead of or in addition to KRAS mutations in early-stage lesions [13]. On the other hand, MCNs tend to accumulate mutations in RNF43 but not GNAS [13,15]. Moreover, unlike PanINs and IPMNs, most MCNs (85%) do not grow within or interact with the main pancreatic duct [13,15,17], likely decreasing the feasibility for developing stool-based screenings. One in three IPMNs and MCNs will become invasive [13], compared with roughly 1% of PanINs [18]. Both IPMNs and MCNs are cystic in nature and larger than PanINs, which makes them easier to detect using abdominal or endoscopic ultrasound, whereas PanINs are solid tumors and are too small to be detected using these methods [13].
328
Theranostic Approach for Pancreatic Cancer
Desmoplastic reaction Pancreatic tumors rely heavily on generating a niche microenvironment to maintain disease progression. Desmoplasia is the mechanism by which PDAC achieves this, and consists of a complex paracrine signaling scheme that takes place between the tumor cells, tumor-associated macrophages (TAM), neutrophils, regulatory T cells (Treg), and pancreatic stellate cells that promotes the carcinogenesis, mutagenesis, and immune evasion of the tumor cells and directly contributes to the infiltrative and metastatic potential of the tumor [27e29]. Both primary and metastatic PDACs exhibit significant desmoplasia and immune cell infiltration from an early stage, and a higher level of extracellular matrix (ECM) deposition in these tumors is associated with a negative prognosis [27,30]. The process starts when pancreatic stellate cells, which are normally quiescent, are induced to differentiate into an activated myofibroblast-like state and proliferate substantially in part as a result of aberrant KRAS and Sonic Hedgehog (SHH) signaling from early cancer cells [7,31] and later by the secretion of growth factors such as platelet-derived growth factor and reactive oxygen species (ROS) from TAM and neutrophils, respectively [27e29]. These activated stellate cells are then stimulated by growth factors secreted from TAMs and neutrophils, including TGFb, fibroblast growth factor, and connective tissue growth factor, to deposit a variety of ECM components such as collagen, hyaluronic acid, osteonectin/ secreted protein acidic and rich in cysteine (SPARC), and other glycosaminoglycans [28,29]. This produces a dense, fibrous, and hypovascularized stroma that provides the perfect environment for carcinogenesis while resisting autoimmunity and chemotherapeutics [27e29]. In addition to activating stellate cells, the secretion of ROS from TAMs and neutrophils has been implicated in multiple aspects of PC, including directly promoting the carcinogenesis and mutagenesis of the cancer cells through inducing DNA strand breaks and base modifications, thereby contributing to the genetic heterogeneity of the cancer cells [28]. Although Tregs do not directly promote carcinogenesis or mutagenesis, they secrete TGFb and suppress autoimmunity, allowing cancer and stellate cells to proliferate [28]. In addition to expressing cell surface and secreted TGFb, some additional markers for tumoral Tregs have been identified, such as CD25 and FoxP3, which are not expressed on other CD4 T cells [28,32]. Taken together, these cancer and immune cells work synergistically to hijack the stellate cells to promote carcinogenesis
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
329
and eventual metastasis of the tumor by generating this niche microenvironment, which represents a major hurdle in the development of novel theranostics [27e29,33].
Molecular markers for pancreatic cancer and their significance in diagnosis and treatment Substantial progress has been made in identifying potential molecular markers to diagnose and treat PC. These markers can be classified into three major classes based on the biological source: serum markers, tumor tissue markers, and cancer stem cell (CSC) markers (Fig. 16.1). We discuss these three types of molecular markers and their importance in theranostics for PC in this chapter. Table 16.1 briefly summarizes the current status of molecular markers and therapeutic targets of PC.
Serum markers Cancer antigen 19.9 Cancer antigen 19.9 (CA 19.9), or sialyl Lewis A, is the best-known and only Food and Drug Administration (FDA)-approved biomarker for monitoring the disease progression and treatment response of PC [34]. Sialyl Lewis A is an aberrant form of disialyl Lewis A, which is normally expressed in pancreatic epithelial cells and participates in immunosuppression [35]. Sialyl Lewis A originates as a result of the epigenetic silencing of a-2,6-sialyltransferase and concurrent hypoxia-induced activation of sialyl Lewis A synthesis enzymes; it is highly overexpressed, accumulated, and secreted in pancreatic and other gastrointestinal tumors [35]. Because CA19.9 is not specific to PC, it cannot be used alone to diagnose PC [34]. However, it has shown some possible diagnostic ability when used in conjunction with other serum markers such as carcinoembryonic antigen (CEA) [36e38], MUC5AC [39], and others. Some antibodies have been developed to target CA19.9, including human monoclonal antibodies (mAbs) 5B1 and 7E3, which demonstrated complement-dependent and antibody-dependent cell-mediated cytotoxicity [34,40,41]. Other antibodies and antibody fragments have been developed for diagnostic, computed tomography (CT), positron emission tomography (PET), and fluorescent visualization applications, including anti-CA19-9 scFv-Fc H310A [42]; an anti-CA19.9 diabody [43,44]; and three 5B1 derivatives, DFO-5B1, FL-5B1, and dual-5B1 [45].
330
Theranostic Approach for Pancreatic Cancer
Molecular Markers and Therapeuc Targets for PC
Serum Markers
Tumor Markers
Cancer Stem Cell Markers
CA19.9
Wilms Tumor 1
CD44
CEA
SPARC
CD24
Mesothelin
VISTA
CD133
IL-6
Hyaluronic Acid
MIC-1/GDF15
Mucins 1, 2, 4, 5AC, 16
Tissue Factor
CD40
Survivin
Her2
TAG-72
Syndecan-1
Osteoponn
Claudin 18.2
Figure 16.1 Molecular markers and therapeutic targets for pancreatic cancer (PC). Many markers are being explored for potential diagnostic and therapeutic applications against PC. Some are expressed on the surface of PC tumor cells and then shed into the serum and could be useful for both detection and tissue-specific targeting, such as mesothelin, tumor-associated glycoprotein 72 (TAG-72), carcinoembryonic antigen (CEA), or cancer antigen (CA)19.9. Others are being targeted with monoclonal antibodyebased immunotherapy, such as the immune checkpoint V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA), which was implicated in PC; or with cancer vaccines and engineered T cells, such as survivin and Wilms tumor 1. Still others are useful targets for stromal depletion therapies, such as CD40 and hyaluronic acid, or for targeted delivery of drugs or visualization agents, such as secreted protein acidic and rich in cysteine (SPARC) and CD44. Her2, human epidermal growth factor receptor 2; IL, interleukin; MIC-1, macrophage inhibitory cytokine 1.
Carcinoembryonic antigen Carcinoembryonic antigen (CEA) is a glycoprotein expressed on the luminal surface of pancreatic epithelial cells and participates in cell adhesion and signaling [34]. CEA has been identified as a potential prognostic
Table 16.1 Molecular markers for diagnosis and therapeutic targets. Target Antibody/agent
CA19.9
Carcinoembryonic antigen
5B1 and 7E3 Anti-CA 19-9 scFv-Fc H310A Anti-CA19.9 diabody DFO-5B1 FL-5B1 Dual-5B1 Labetuzumab
KAb201
Mesothelin
M5A T84.66 Amatuximab Anetumab ravtansine DMOT4039A SS1P (Fv fragment) LMB-100 (Fab fragment) CRS-207 (Listeria live attenuated vaccine) JNJ-64041757 (Listeria live attenuated vaccine) CAR T treatments (various)
Utilization
Source
Therapeutic Diagnostic Diagnostic Diagnostic Diagnostic Diagnostic Therapeutic, diagnostic Drug conjugation Radioimmunotherapy conjugation Radioimmunotherapy conjugation Immunofluorescence Immunofluorescence Therapeutic Therapeutic Drug conjugation Drug conjugation Drug conjugation Drug conjugation Vaccine Vaccine
[34,40,41] [42] [43,44] [45] [45] [45] [46,47] [47e49] [50]
[52,53] [54] [55e58] [60,61] [62] [59,64] [59,65] [59,66] [59,67] [59,68]
Therapeutic
[59,69e73]
[51]
Continued
Table 16.1 Molecular markers for diagnosis and therapeutic targets.dcont'd Target
Antibody/agent
Utilization
Source
MUC1
1B2 12D10 Anti-hMUC1 PankoMab Clivatuzumab Antibodies (various) Vaccines (various) Vaccines (various) B72.3 Indium-111 satumomab pendetide 3E8.scFv.Cys-IR800 3E8 (124)I-mCC49 CAR T treatments (various) Zolbetuximab Claudiximab CAR T treatments (various) LY2181308 (antisense oligonucleotide) YM155 (inhibitor) Survivin-2B80-88 (peptide vaccine) Dendritic cell vaccine Engineered cytotoxic T cells Trastuzumab
Diagnostic Diagnostic Diagnostic Therapeutic Therapeutic, diagnostic Diagnostic Therapeutic Therapeutic Therapeutic, diagnostic Imaging Imaging Therapeutic, diagnostic Imaging Therapeutic Therapeutic Therapeutic Therapeutic Therapeutic Therapeutic Therapeutic Therapeutic Therapeutic Therapeutic
[179] [179] [180,181] [182e184] [185e187] [188e191] [170,188,192,193] [194e197] [101] [101] [103] [104] [105] [106] [212e217] [212,213] [218e221] [92e94] [95] [96] [97] [98] [205]
Siltuximab Clazakizumab sgp130Fc (soluble IL-6R analog) Various additional mAbs
Therapeutic Therapeutic Therapeutic
[123] [126] [127] [128]
MUC5AC MUC4 MUC2 TAG72
Claudin 18.2
Survivin
Human epidermal growth factor receptor 2 IL-6
IL-6R Tissue factor
Osteopontin Secreted protein acidic and rich in cysteine CD44
V-domain immunoglobulincontaining suppressor of T-cell activation Hyaluronic acid CD40
Tocilizumab Sarilumab (89)Zr-Df-ALT-836 64Cu-FVII 1849-ICG (111)In antietissue factor 1849 CAR T treatment HuMax-TF-ADC hu1A12 Osteopontin-VEGF bispecific mAb Abraxane (paclitaxel-conjugated albumin) Paclitaxel/curcumin double-tagged albumin
Therapeutic Therapeutic Imaging Imaging Immunofluorescence Diagnostic Therapeutic Drug conjugation Therapeutic Therapeutic Drug conjugation Drug conjugation
[124] [125] [85] [86] [87] [88] [89] [90] [111] [112] [139] [141e145]
Multiple HA-BODIPY HA-40 -(aminomethyl)fluorescein HA-2,3,5-triiodobenzoic acid HA- tris(hydroxypyridinone) amine JNJ-61610588
Drug conjugation Imaging Imaging Imaging Imaging Therapeutic
[237e245] [238] [245] [244] [245] [153]
PEGPH20 CP-870,893 Chi Lob 7/4
Stromal depletion Therapeutic Therapeutic
[158e168] [201,202] [202,203]
CA, cancer antigen; CAR, chimeric antigen receptor; HA, hyaluronic acid; IL, interleukin; mAb, monoclonal antibody; MUC, mucin.
334
Theranostic Approach for Pancreatic Cancer
indicator during chemotherapeutic treatments because the serum level of CEA is directly associated with the progression of tumor invasion and malignancy [37] and increases the specificity of the diagnosis of PC when used in combination with CA19.9 [36e38]. Some clinically relevant antibodies have been generated against CEA. First, labetuzumab is a humanized mAb that shows diagnostic and therapeutic potential and is currently undergoing clinical trials [46,47]. It has been studied in the context of antibodyedrug conjugates [47e49] and was used in radioimmunotherapy conjugation [50]. KAb201 is another humanized anti-CEA antibody that has similarly high pancreatic tumor specificity and was investigated for the delivery of radioisotopes [51] and conjugation to fluorophores for development in immunofluorescenceguided surgery [52,53]. A third antibody, M5A, is being developed for fluorophore conjugation [54]; a different version of that antibody, T84.66, has been humanized [55] and is undergoing clinical trials [56e58]. Mesothelin Mesothelin is a cell surface molecule that is highly overexpressed in PC [59,60]. Its purpose is unclear but is thought to participate in cell adhesion and binding to CA125 and MUC16 in mesothelial cells and aids metastasis in many cancers [59]. Many mesothelin-targeting treatments are under development [59]. Amatuximab is one such chimeric mAb that is in a phase II clinical trial and interferes with mesothelin’s binding to CA125 and MUC16 [60,61]. There are also many antibody-drug conjugates in development: anetumab ravtansine is in phase I and II clinical trials [62] and consists of a human mAb that delivers the linked microtubule-targeting toxin DM4 to the cancer cell, which internalizes it and secretes a metabolite that in turn kills surrounding cells [63]. DMOT4039A is another antibody-drug conjugate that delivers the antimitotic monomethyl auristatin E to target cells; it has been tested in PC patients [59,64]. SS1P is an Fv antibody fragment that delivers Pseudomonas exotoxin A to mesothelin, which is endocytosed and targets eIF-2a [59,65]. Similarly, LMB-100 is an Fab-PE24 immunotoxin that is entering clinical trials [59,66]. Other therapies include Listeria live attenuated vaccines CRS-207 [59,67] and JNJ-64041757 [59,68]; and various chimeric antigen receptor (CAR) T therapies [59,69e73]. Macrophage inhibitory cytokine 1 Macrophage inhibitory cytokine 1 (MIC-1), also known as growth differentiation factor 15, is a secretory growth factor that is implicated in many
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
335
aspects of tumor invasion, including migration [74] and immunosuppression [75], and is overexpressed within tumors and serum of PDAC patients [76e79]. MIC-1 is of particular interest because of its ability to detect earlystage PDAC before CA19.9 is detectable [76,78], and in the roughly 10% of the patients who do not express CA19.9 at all [78,79]. It can differentiate between early-stage PDAC and IPMN [80] and was able to distinguish between PDAC and pancreatitis when used in conjunction with UL16 binding protein 2 [76] or CA125 [81], but not when MIC-1 was used alone [77]. Therefore, MIC-1 is an important marker for early detection and monitoring of PC and warrants further investigation. Tissue factor Tissue factor (TF) is a cell surface receptor normally expressed in subendothelial cells that binds soluble Factor VII (FVII) upon rupture of the blood vessel to initiate clotting rapidly [82]. It is also aberrantly expressed in PDAC tumor cells, which use its secondary effect of activating proteinaseactivated receptor-2-integrin signaling to initiate angiogenesis, but with the side effect of triggering clotting and thrombosis within the tumor and subsequently causing thromboembolisms at distant sites within the body [83]. Because the expression within PDAC tumors is correlated with disease progression [84] and the increased occurrence of thromboembolic comorbidities in PC patients [83], TF is a promising target for both therapeutics and diagnostics with many theranostic applications being developed. For example, TF has been explored as a target in PET imaging using both radiolabeled anti-TF antibodies [85] and FVII ligand [86]. Other applications targeting TF include immunofluorescence [87], an anti-TF application of immune-SPECT/CT [88], a TF-CAR T cell [89], and an antibody-drug conjugate that showed regression of patient-derived xenografts with minimal off-target effects [90], among others. Survivin Survivin is an antiapoptotic protein normally expressed in embryonic and fetal tissues but not in adult tissues [91]. It was found to be expressed in 81% of PDAC tumors; elevated serum concentrations were associated with metastasis and invasiveness, but not the size or location of the tumor [91,92]. Because survivin is expressed in tumors as well as in the serum, it is unique in that it could serve both as a therapeutic target and diagnostic marker for PDAC. Many attempts at targeting survivin have not panned out, such as with antisense oligonucleotide LY2181308 [92e94] and
336
Theranostic Approach for Pancreatic Cancer
inhibitor YM155 [95], but trials involving survivin, peptide vaccines have seen greater success, with prolonged survival and disease stabilization in one phase I trial of the survivin-2B80-88 peptide vaccine [96]. Another cancer vaccine trial is also being attempted using peptide-pulsed dendritic cells [97]. In addition, a study involving engineered cytotoxic T cells targeting survivin is currently under way [98]. Tumor-associated glycoprotein 72 (TAG72) TAG72 is a cell surface glycoprotein that functions similar to an MUC and is often overexpressed in and shed from PC [99]. It is known to be a better prognostic indicator than CA19.9 or CEA [100]. Despite this and the fact that Indium-111 satumomab pendetide, a derivative of the anti-TAG72 antibody B72.3, was the first monoclonal antibody to be approved for tumor imaging [101], other attempted applications of anti-TAG72 mAbs fizzled out. Luckily, new attempts are being made to target TAG72 for prognostic evaluation [102], immunofluorescence-guided surgery [103], the development of new antibodies such as 3E8 [104], improved targeting in PET and CT [105], and CAR T treatments [106]. Osteopontin Osteopontin is an ECM glycoprotein secreted in PDAC; it is implicated in PDAC disease progression. It is expressed as one of three splice variants in PDAC, with osteopontin-a expressed in nearly all PDAC, osteopontin-b expression correlating with survival, and osteopontin-c correlating with metastatic disease [107]. Because PDAC secretes alternatively spliced forms of osteopontin, it shows potential for tumor- and disease stage-specific targeting. Although the exact mechanisms of osteopontin signaling in PDAC are unknown, it binds to CD44 and integrins to trigger processes such as tumor progression and complement inhibition [108]. Osteopontin also drives metastasis by triggering the release of vascular endothelial growth factor (VEGF) and matrix metalloprotease (MMP), which is inhibited by knocking down osteopontin [109]. This process is also stimulated by nicotine, which is the proposed mechanism by which smokers experience elevated PC risk [109]. Osteopontin is being explored as a marker for PC. It was found to perform better than CA19.9 in discerning IPMN [80] and resectable PDAC from pancreatitis [110]. Antiosteopontin antibodies are being developed, including hu1A12, which inhibited metastasis in an in vivo study [111] and also when hybridized with the anti-VEGF antibody bevacizumab [112]. At least one clinical trial is exploring the use of
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
337
osteopontin as a marker of intratumoral hypoxia [113]. However, this marker remains relatively unexplored. Interleukins 6, 8, and 10 Feng, et al. and others showed a correlation between high serum levels of interleukins (ILs) 6, 8, and 10 and poor prognosis of PC [114e116]. IL-6 and IL-1b have similarly been shown to indicate prognosis of PC patients undergoing gemcitabine therapy [117]. IL-6 and IL-8 are proinflammatory cytokines expressed by many PDAC tumoral cell types; they are required for PDAC progression, metastasis, and angiogenesis [118,119]. IL-10 is expressed by Tregs within the tumor and is important for cell proliferation and immune suppression [120,121]. These cytokines work synergistically to promote PDAC disease progression and avoid autoimmune responses [114,122]. They are an attractive target for both therapeutics and diagnostics because they are both expressed within the tumor and also secreted into the serum. Three IL-6 or IL-6R antibodies have been approved: siltuximab, tocilizumab, and sarilumab. A clinical trial using siltuximab, an anti-IL-6 chimeric mAb, against solid tumors including PC proved inefficacious [123]. Tocilizumab, which is a humanized anti-IL-6R mAb, is undergoing a clinical trial for PDAC [124]. Sarilumab, an anti-IL-6R human mAb, has shown efficacy against solid tumors in preclinical trials [125] but has not yet been tested against PC. Other anti-IL-6 antibodies are in development, such as clazakizumab, an anti-IL-6 human mAb [126], and sgp130Fc, a soluble IL-6R analog that inhibits IL-6 trans-signaling [127]. Many other IL-6 antibodies are in various stages of development and are reviewed here [128].
Tumor tissue markers Wilms tumor 1 antigen Wilms tumor 1 antigen (WT1) is expressed in around 75% of PDAC tumors but is absent from normal pancreatic ductal epithelium [129]. Knockdown of WT1 impairs tumor growth in vitro, which indicates that it has an unknown role in PDAC pathogenesis [129]. However, no association has been made between WT1 expression and stage or location of the tumor, which makes it a better target than prognostic indicator [129]. WT1 has been identified as the number one target for developing cancer vaccines against PC by the National Cancer Institute [130,131], and is the subject of multiple cancer vaccine [132,133] and CAR T clinical trials [134,135].
338
Theranostic Approach for Pancreatic Cancer
Secreted protein acidic and rich in cysteine (SPARC) Pancreatic stellate cells maintain the dense stroma in PDAC partially through constant remodeling of the ECM, which requires deposition of new collagen and organization of fibronectin fibrils [136]. SPARC expression from these stellate cells drives aberrant ECM remodeling in PDAC in part through stimulating expression of MMP [137]; it is also involved in tumor invasiveness [136]. SPARC is being investigated as a means of targeting drugs to PDAC tumors because of its high affinity for binding albumin [138]. One such drug conjugate, Abraxane or nabpaclitaxel, was approved by the FDA in 2005 [139]. Applications of this technology targeting PC cancer include coadministration with gemcitabine [140], a paclitaxelecurcumin double-tagged albumin [141], along with other methods of complexing and delivering these agents [142,143]. Studies demonstrated the efficacy of this approach, showing significant improvements in response and survival [144,145]. Another group approached this method from an even more innovative direction by incorporating a maleimide group into the drugs, creating prodrugs that readily conjugated with albumin in vivo [146]. At least one other clinical trial using albuminconjugated paclitaxel is ongoing [147]. Overall, these successes demonstrating the targeting of SPARC within PDAC tumors represent an avenue of therapeutic design that should be explored further. V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA) Immune checkpoint inhibitors have yielded tremendous success in other cancers but have seen little success in PC [148]. The coinhibitory receptor PD-1 is expressed on immune cells and normally binds to programmed death (PD) ligand 1 (PD-L1) expressed on host cells to prevent autoimmunity [149], but although it is commonly upregulated in other tumor cells as a method of immune evasion, it is nearly absent from PCs owing to the lack of T-cell infiltration, rendering current PD1/PD-L1 treatments ineffective [148,149]. On the other hand, pancreatic tumors express CTLA-4 on Treg cells [150], which rely on its coinhibitory function to evade the antitumor immune response. Despite the expression of this immune checkpoint, however, anti-CTLA-4 antibodies have proven ineffective against PC tumors [150]. Blando et al. discovered that this is because a second immune checkpoint, V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA), is expressed on the surface of CD68þ TAMs within PC tumors, which enables them to bypass single-agent
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
339
anti-CTLA-4 therapies [151,152]. Janssen Pharmaceuticals is developing an anti-VISTA antibody, JNJ-61610588, which is under clinical trial [153], and more treatments targeting VISTA are sure to come. This discovery may soon revolutionize the way immune checkpoint therapy is pursued in PC. Hyaluronic acid Hyaluronic acid (HA) is an important ECM component that is crucial to PDAC pathogenesis. HA is secreted from both tumor and stromal cells [154] and is a long glycosaminoglycan consisting of numerous disaccharide repeats [155]. It is one of the ECM components responsible for the high interstitial pressure within the PDAC tumor, because it draws huge amounts of water (roughly 15 molecules per disaccharide) into the tumor, resulting in subsequent blood vessel collapse [155,156]. The level of HA content within PDAC tumors is also associated with epithelial to mesenchymal transition (EMT) and poor prognosis and survival [27,157]. HA has been evaluated for its ability to target and deplete the PDAC stroma [158]. After an initial half-life of 3 min, PEGPH20 was engineered for greater therapeutic efficiency via PEGylation of the human hyaluronidase PH20, which extended its half-life to over 10 h [158]. Many clinical trials have been conducted using PEGPH20 in combination with existing chemotherapeutics, such as dexamethasone or oxaliplatin, irinotecan, fluorouracil, and leucovorin, and many more are under way [159e165]. In general, these trials have shown substantial benefits including decreases in tumor HA level and increased drug perfusion [166]. However, there are significant incidences of thromboembolism in multiple study participants [167,168], which it is hoped may be overcome in subsequent trials. Mucins 1, 2, 4, 5AC, and 16 MUCs are glycoproteins that form a protective layer on normal epithelial cells and perform a variety of tasks that are exploited in PC [169,170]. MUCs contain a tandem repeat polypeptide backbone and up to 90% by weight of N-acetyl-galactosamine oligosaccharide chains branching from the main peptide [170]. Of importance to PDAC, MUC1 and MUC4 are primarily membrane-bound, whereas MUC2, MUC5AC, and MUC16 are secretory [170]. Mucins aid in immunosuppression and metastasis by PDAC tumors in a variety of ways [169e174]. For example, MUC1 and MUC16 can block antigen presentation through binding to siglecs and Toll-like receptors [169]. Overexpression of MUCs can also block cell surface antigens, or they can induce anergy on T cells through binding
340
Theranostic Approach for Pancreatic Cancer
ICAM-1 [169]. MUC4 also appears to produce a splice variant that aberrantly activates the integrin-b1/FAK/ERK pathway to participate directly in the metastasis of PDAC [175]. MUCs are differentially expressed in PanINs, IPMNs, and invasive PDACs, and thus offer the potential to differentiate these lesions at the molecular level [13]. Of these MUCs, only MUC1 is expressed in normal pancreatic ductal epithelium, but is highly overexpressed in PanIN-2/3 and PDAC, which makes it a popular target among emerging therapies [170]. MUC2 is not expressed on PanINs or PDAC but are in IPMN, which could be exploited for the molecular differentiation of these lesions [13]. MUC4, MUC5AC, and MUC16 (as well as MUC3) are expressed increasingly as PanIN progresses from early to infiltrative lesions and are highly expressed in PDAC; they could be useful in staging these tumors [13]. It was shown that combining MUC5AC with CA19.9 screening improved the accuracy of diagnosing resectable PC and allowed for differentiation among PC, pancreatitis, and benign lesions [39]. Further exploiting these expression differences in the precursors of PDAC will be crucial to the development of diagnostics and therapeutics in the near future. Given their importance to the pathogenesis of multiple cancers and the frequent occurrence of tumor-specific neoantigens in these glycoproteins [169e174], MUCs are the target of many therapeutics in development. Early attempts in both antibody and vaccine development failed because most of them recognized the polypeptide structure of the glycoproteins rather than the glycosylations, or because the antigens used to produce them had patterns of glycosylation different from those of the cancers they intended to target [170,176]. The importance of tumor-specific glycosylations has since become more clear [170,177], and the increasing capability to generate tumor-specific anti-MUC mAbs and the possibility of better mapping the epitopes of new antibodies using glycopeptide libraries [178,179] has allowed for greater characterization and exploitation of MUC tumor-specific antibodies. Two such antibodies, 1B2 and 12D10, were engineered using glycopeptide libraries to possess specificities for both glycan and peptide sequences [179]. Another antibody, anti-hMUC1, was found to bind to pancreatic tumor-specific MUC1 while avoiding MUC1 expressed on normal tissues in vivo. It was found to be internalized by the PC cells [180], opening up the potential for further developing a toxin- or radioisotopeconjugated version of this antibody for therapeutic or diagnostic use, an
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
341
area in which previous attempts have failed [181]. PankoMab is another MUC1-reactive antibody designed to bind preferentially to tumorassociated MUC1 [182]; it is being evaluated in phase I and II trials [183,184]. Clivatuzumab is an anti-MUC5AC antibody in clinical trials that is reactive against PanINs and PDAC. It shows promise for use as a serum biomarker [185e187]. MUC5AC is a secretory MUC that is not expressed in normal pancreatic epithelia, but it is expressed in early-grade PanINs, which makes this antibody one of the most promising in allowing early biochemical detection of precancerous lesions of the pancreas [185e187]. MUC4 remains relatively unexplored in PC therapies, but some preliminary investigations into antibodies [188e191] and vaccines [170,188,192,193] are under way. MUC2 also has not been widely explored other than a series of vaccine clinical trials for prostate cancer [194e197], but it could be useful in the early detection of IPMNs [13]. CD40 Normally, helper T cells should activate macrophages to attack cancer cells by stimulating CD40 on the macrophage surface [198]. However, this process does not normally occur in PDAC owing to the prevalence of immune suppressive cells such as Tregs, tumor-associated macrophages, and myeloid-derived suppressor cells, which results in an immunosuppressive tumor environment and subsequent lack of effector T-cell infiltration or activation [30,198]. Beatty et al. were able to demonstrate that stimulating CD40 activation using an agonist antibody resulted in stromal depletion and tumor regression by means of noneT cellemediated macrophage infiltration [198]. Since then, others have explored the use of CD40 agonists and found that they are capable of producing stromal depletion through both T celledependent andeindependent responses against PDAC tumors [199]. Multiple CD40 agonist antibodies are under development [200], including human mAb CP-870,893 [201,202] and chimeric mAb Chi Lob 7/4 [203], but so far they have shown limited efficacy in clinical trials [202,203], and remain an area of active investigation. Human epidermal growth factor receptor 2 (Her2) Human epidermal growth factor receptor 2 (Her2) has garnered particular interest because anti-Her2 therapies are already on the market with more in development. This epidermal growth factor receptor is not expressed on normal pancreatic ductal epithelium but is found in some PanINs and
342
Theranostic Approach for Pancreatic Cancer
PDAC, although the prevalence and extent are debated. Studies have reported Her2 overexpression in as little as 2% [204] to 11% [205], 45% [206], and nearly 70% [16,207] of patient samples. Results from clinical trials involving anti-Her2 inhibitors and mAb have also been hit or miss [208]. One drug, trastuzumab, targets only highly overexpressed Her2, and although Her2 was expressed in 60% of PDAC tumors in one trial, only 11% were deemed to be adequately overexpressed to the extent required for the treatment to be successful [205]. Many other anti-Her2 therapeutics are still under development; whether this receptor can be successfully targeted in PC remains to be determined. Syndecan-1 PDAC cells rely heavily on the mass import of extracellular nutrients through macropinocytosis to sustain their growth [209]. It was discovered that syndecan-1, an important mediator of PDAC macropinocytosis, is expressed on the surface of PDAC tumor cells as a downstream response to mutant KRAS [210]. Syndecan-1 is a heparan sulfate proteoglycan that was previously shown to mediate macropinocytosis, but it is now known that this surface marker is upregulated starting early in PC disease progression and is found in pancreatitis and premalignant PanIN lesions [210]. The same study also showed that inhibition of syndecan-1 resulted in suppressed tumor growth, which indicates its potential utility as a drug target [210]. Although the clinical utility of syndecan-1 and macropinocytosis in treating PDAC remains unknown, it will undoubtedly be an area of intense interest in the future. Claudin 18.2 Claudin 18.2 is an emerging target for PDAC. It was shown to be aberrantly expressed in 59% of tumor specimens but not in normal pancreatic epithelia [211]. Claudin 18.2 is a gastric epithelial tight junction protein normally expressed on luminal regions of polarized cells, which should offer minimal exposure to antibody-based therapies [211,212]. Zolbetuximab, also known as IMAB362 or claudiximab, is an anti-Claudin 18.2 antibody entering phase III clinical trials [212]. It has been shown to target PDAC cancer cells and induce cytotoxicity through antibody-dependent and complement-dependent pathways [213]. Trials using zolbetuximab combined with existing chemotherapeutics are ongoing [214e217]. Many CAR T Claudin 18.2 trials are also enrolling [218e221].
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
343
Cancer stem cell markers Although they make up only around 1% of cells within the tumor and their origin is still not completely understood, CSCs are increasingly recognized as a key driver of metastasis, drug resistance, and disease recurrence in PDAC [222e224]. Since their discovery in 2007 [225,226], PC stem cells (PSCS) rely on multiple common developmental pathways such as SHH, Wnt, Notch, PTEN, and Bmi-1 [222,223]. The prevalence of these developmental pathways in CSCs of many origins has prompted their targeting with various drugs, but the cells frequently compensate for the loss of one of these pathways by upregulating another and quickly develop resistance to these inhibitors [224]. It has also been reported that gemcitabine promotes its own resistance by inducing stemness in PC cells [227e229]. Many groups have shown efficacy in vitro by targeting some of these developmental pathways [222,230], but it remains to be seen whether these repurposed drugs or other efforts to reengineer some chemotherapeutics to stave off resistance will pan out [222,223,231e234]. CD44, CD24, and CD133 As with other stem cells, PCSCs are self-renewing and can produce multiple lineages of differentiated progeny [222e224]. Early characterization in vitro fell along similar lines as with induced pluripotent stem cells in that CSCs can form spherical clusters in low-adhesion conditions and can seed new tumors when implanted [222,223]. Several surface markers have since been identified, such as CD24, CD44, CD133, epithelial-specific antigen, c-Met, aldehyde dehydrogenase (ALDH), and C-X-C chemokine receptor type 4, which are associated with stemness and gemcitabine resistance in PCSCs [222e224]. Despite the usefulness in these markers for new applications such as microfluidic chip liquid biopsy [235], these markers are specific to many cell types [236] and it remains to be seen whether they will be useful in targeted drug delivery. CD44 is a membrane glycoprotein that is associated with PC cells, specifically PCSCs [237]. CD44 expression is associated with poor prognosis and radiation therapy resistance and is expressed in two isoforms: CD44s and CD44v [237]. CD44v is associated with metastasis, and isoform switching between s- and v-forms has also been observed with EMT [237]. Aside from being a marker for PCSCs as discussed earlier, CD44 has also been explored extensively for targeted drug delivery [237e245] and tumor visualization, including HA-BODIPY [238] and HA-40 -(aminomethyl)
344
Theranostic Approach for Pancreatic Cancer
fluorescein [245] for confocal imaging, an HA-2,3,5-triiodobenzoic acid conjugate used for fluorescence and CT imaging [244], and HAtris(hydroxypyridinone)amine for SPECT/CT [245]. HA has also been conjugated to gemcitabine-containing liposomes for targeting CSCs [246]. As noted earlier, a plethora of HAedrug conjugates are under development.
Current diagnostics Once PC is suspected, a variety of noninvasive or minimally invasive diagnostic procedures may be used to locate and stage the tumor [3e5,7,247]. CT and MRI are recommended as the first modes of visualization because both offer fine resolution of the surrounding tissues with similar accuracy [5,7,248,249]. However, CT is considered inadequate for determining tumor resectability because of its low sensitivity in detecting small metastases, which must instead be characterized laparoscopically [250]. The use of MRI has increased over time, owing to the inherent advantage of not exposing the patient to ionizing radiation as in CT [2,7]. MRI is superior in its resolution of soft tissue and the ability to visualize simultaneously the ducts, vasculature, liver, and peritoneum, in which metastases are common [5,250,251]. Abdominal ultrasound is also commonly used as a precursor to CT because it is noninvasive, available, and affordable and can differentially diagnose conditions with similar symptoms such as gallstones and ulcers while avoiding the use of ionizing radiation [2,5,21]. However, because the pancreas lies deep within the abdominal cavity, abdominal ultrasound cannot easily visualize small lesions or discern between benign and metastatic tumors [2,21]. Endoscopic ultrasound is widely used upon initial diagnosis of PDAC because of its high sensitivity and accuracy compared with MRI and CT, especially in diagnosing small, premetastatic, early-stage tumors, and because of its useful applications such as stenting, endoscopic fine needle aspiration, and endoscopic retrograde cholangiopancreatography [7,249]. PET can also be useful in staging and diagnosis because of the ability to discern between benign and metastatic lesions by measuring the rate of glucose uptake through the use of 18F-fluorodeoxyglucose [252] and many PC-specific markers are under development [42e45, 85,86,88,105,247,250,253].
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
345
Emerging diagnostics Despite the usefulness of current diagnostic techniques in identifying laterstage tumors, their high cost and invasiveness or risk for comorbidities prevent their use in preemptive screening for PC in those without a genetic predisposition [2]. Currently no approved serum molecular markers can be used to diagnosis PC [3e5,7]. Although the serum marker CA19-9 is commonly used to determine the prognosis of PCs during therapeutic regimens, it is not specific to PC and cannot be used for initial diagnosis [5,7,37,254]. Identifying minimally invasive, reliable, and affordable screening methods is crucial to reducing the burden of PC. The focus on developing liquid biopsies for various cancers has yielded some promising results, and these advances are moving the reliable early detection of PC closer to reality.
Blood-based DNA screening The development of a routine blood-based PC screen would revolutionize this field, but little progress had been made until recently. An expansive study in 2014 by Bettegowda et al. found that circulating tumor DNA was detectable in most patients with advanced PC and 48% of patients with localized tumors [255]. They also found that 87% of patients that had KRAS mutant tumors could be identified using circulating tumor DNA [255]. This cell-free circulating DNA is released into the bloodstream by dying cancer cells, and because most cancers have at least one trademark somatic mutation [256], the detection of cancer-specific alleles diluted by wild-type DNA in the blood can be accomplished using polymerase chain reaction (PCR)-based methods [255,257,258]. Two subsequent studies by Cohen et al. focused on detecting early-stage (stages IeIII) cancers using circulating tumor DNA, culminating in the development of the CancerSEEK PCR panel [257,258]. CancerSEEK uses 61 sites within 16 common cancer genes to identify a wide range of cancers. It is innovative in its additional combination of protein markers and machine learning to predict the tissue of origin of the cancer accurately [257,258]. It is estimated that this panel could detect up to 95% of PC [258]. This ability to detect premetastatic PC accurately is unprecedented, and the possibility of combining other markers such as cancer-specific promoter methylation offers the possibility to expand and refine this technology [256,259].
346
Theranostic Approach for Pancreatic Cancer
Marker panels To date, numerous markers have been identified that can detect early lesions, track disease progression, and differentiate among PDAC, PanIN, IPMN, and benign lesions, pancreatitis, and nonpancreatic diseases [79]. Several serum panels have been announced that will target some of these markers. The first is an antibody microarray that targets 29 markers including Cdk2, IL-6, VEGF, and sialyl Lewis X, and had an accuracy of 96% in detecting early-stage (stage I and II) PDAC [260]. The second is a sandwich enzyme-linked immunosorbent assayebased assay of three markers, CA19.9, MUC5AC, and MUC16, to detect stage I and II cancers with 75% accuracy and only a 5% false-positive rate [261]. Finally, the third is under development by the GRAIL company, which hopes to develop a liquid biopsy panel capable of detecting multiple cancers [262]. PC marker panel development remains an active area of research; more such panels are sure to come.
Cell-based screening The use of microfluidic devices has revolutionized the isolation of circulating tumor cells (CTCs). Previous attempts at detecting CTCs proved difficult because these cells are heavily diluted by circulating blood cells and are also prone to shearing, anoikis, and attack by immune cells [263]. Because of their high surface area and the ability to coat them with an antibody of choice, rather than pretagging the CTCs within the sample, microfluidic chips have become increasingly useful in isolating CTCs [264,265]. This technology has been used to isolate and then successfully culture CTCs [264] and to isolate CTCs and circulating PCSCs from patient samples [235], which shows that it is gentle enough to isolate cells without destroying them, allowing for further use of biochemical assays downstream. It can also be specific enough to distinguish between similar cell types, which would be important for applying this technology to staging diseases. Many variations are also being studied that integrate various physical methods to maximize contact with the chip surface and methods of mechanical sorting to increase the efficiency and purity of the isolated cells [266e268]. Although this technology is still early, the development of cellbased screening using microfluidic chips show great potential for the early diagnosis of PCs as well as improved monitoring of disease progression and response to treatments.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
347
Stool-based DNA screening Developing a noninvasive and cost-effective procedure for detecting PDAC and invasive pancreatic lesions is the holy grail of PC diagnostics. Success in detecting other gastrointestinal cancers through stool-based DNA screening has encouraged a similar search for genetic targets in the screening of PC [269]. Early attempts at detecting KRAS and P53, the two most common genetic alterations in PC, in stool samples of PDAC patients were not sensitive or specific enough to detect PDAC reliably [270]. In a subsequent study, Kisiel et al. succeeded in identifying nine aberrantly methylated genes in PDAC stool samples that could be used to identify PC accurately [271]. Of these, bone morphogenetic protein 3 was the most promising because it was more discriminant than the next best three markers combined and had a sensitivity of 67% when combined with KRAS [271]. A clinical trial investigating both genetic mutations and hypermethylation in stool samples of patients with PC and IPMN is also under way [272]. Ultimately, the proximity of PDAC, PanIN, and IPMN lesions to the pancreatic duct combined with limited options for early detection and tracking disease response to treatment make stool-based detection an important area of research.
MicroRNA screening Compared with to DNA screens, relatively little work has been done to identify a microRNA (miRNA) profile for PC. miRNAs are short, singlestranded RNA fragments that regulate gene expression posttranslationally [273]. Since the discovery that cancers often use miRNAs to regulate protein expression [274], many groups have investigated the usefulness of targeting cancer-specific miRNAs for therapeutic purposes [275]. Ultimately, miRNA-based therapies have been hit or miss, hindered by a lack of tumor-specific delivery, low uptake of the sequence, and the tumor’s ability to adapt quickly to the drug [275]. However, profiling tumorspecific miRNA expression patterns may still be useful for detecting PC. Of many studies identifying miRNA signatures in cancers of the pancreas [273,275e280], a few good candidates have emerged for detection and differentiation of pancreatic lesions and cancers, including miR-103, miR-107, and miR-155 to tell tumors apart from normal pancreatic tissue, miR-204 to detect insulinomas, and miR-21 liver metastasis [276]; miR-1290 to detect early cancers and IPMN lesions [278]; and miR-133a
348
Theranostic Approach for Pancreatic Cancer
to distinguish primary from metastatic tumors [279]. Because of the lack of ability to identify and differentiate easily between early and benign lesions of the pancreas, miRNA profiling may prove to be an important tool in the staging and monitoring of the progression of PC in the near future.
Drug resistance PDAC tumors are heterogeneous in cell type and genetic composition and use a variety of mechanisms for developing and maintaining disease progression. One such mechanism is the PC’s innate and adaptive drug resistance capabilities. The first line of defense is the dense, hypovascularized stromal environment that maintains a high interstitial pressure to resist the perfusion of therapeutics [7,27e29,33]. Next, each tumoral cell type can employ various mechanisms for overcoming the drug, including reducing the mechanism of uptake, relying on hypoxia-induced autophagy to avoid uptake, increasing the efflux of the drug via ABC transporters and nucleoside transporters, inducing senescence in response to cues from the tumor environment, or circumventing the drug’s effects by downregulating the targeted pathway, increasing catabolism of the drug, and increasing expression of antiapoptotic proteins [28,281]. A study by Halbrook et al. showed that TAMs also actively participate in resistance to gemcitabine, a long-standing frontline therapy for PC, by releasing species of pyrimidine metabolites that mimic and outcompete gemcitabine for uptake into the cancer cells [282]. Pancreatic tumors especially rely on the adaptive chemoresistance abilities of CSCs, which either are innately resistant to most current therapeutic regimens or can quickly develop resistance through the same mechanisms as listed earlier [222e224]. They also have an important role in engaging cross-talk between tumoral cell types to initiate EMT, which allows the tumor cells to alter their proteome to resist treatment and promotes posttherapy immune escape, ultimately reseeding tumor colonies once the treatment subsides [222e226,283].
New insights into mechanisms of radioresistance CSCs are also a focus of intense interest in PC because of their ability to resist chemotherapy. Ionizing radiation relies in part on the generation of ROS to induce DNA damage in target cells, but CSCs can resist this generation of ROS through multiple mechanisms. For example, CSCs normally maintain characteristically low levels of ROS; this is known to be mediated in part through the upregulation of free radical scavengers such as
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
349
glutathione [284] and the increased production of reduced nicotinamide adenine dinucleotide phosphate that occurs through an aberrant glutamine pathway driven by KRAS and Nrf2 [285]. However, other mechanisms for radiotherapy resistance are suspected, including increased DNA damage repair. Although the full host of mechanisms is unknown, there have been some advances in our understanding of PCSC radioresistance; these discoveries could inform attempts to develop pretreatments or radiotherapy sensitization therapies for PCSCs in the near future. A report by one group shed light on the subject: ALDH is known to be overexpressed in many CSCs and is associated with both chemotherapeutic and radiotherapeutic resistance [286]. However, although the chemoresistant function of ALDH is understood to rely on its ability to catabolize many different chemotherapeutic drugs, its association with radioresistance is less understood [286,287]. Harati et al. discovered that Nanog regulates ALDH expression in CSCs, and that Nanog also induces postradiation double-stranded break DNA repair by regulating Notch1 and Akt [287]. In other words, an increase in ALDH expression observed by many in radioresistant CSCs actually appears to be evidence of upstream Nanog-Notch1-Akt DNA repair in those cells [287]. This discovery may reveal the mechanism by which coadministration of ALDH inhibitors has been observed to sensitize cells to chemotherapy and radiotherapy [288,289], and may result in more effective strategies for targeting PSCSs in vivo. In another discovery, Tsubouchi et al. demonstrated that x-ray radiation directly upregulates the expression of CD44, which in turn maintains ERK phosphorylation (which is involved in cell proliferation) and drives EMT to promote the adaptation of tumor cell protein expression profiles to become more resistant to the therapy [290]. PCSCs that express CD44 are known to be particularly resistant to radiation therapy. Its expression in PDAC tumors has long been associated with poor prognosis [291]. However, although it was assumed that the increase in CD44-expressing PCSCs after x-ray radiation was caused by the relative depletion of non-CD44 cells, this observation may indicate that radiotherapy directly promotes therapeutic resistance and metastasis [290,292].
Penetration enhancers Many groups are currently developing technologies to enhance the penetration of drugs and imaging agents into tumors, including the use of amphiphilic molecules that increase the efficiency of drug delivery.
350
Theranostic Approach for Pancreatic Cancer
One such technology is the penetration enhancer INT230-6, which is a combinatorial therapy in phase I/II clinical trial targeting PDAC [293]. INT230-6 is composed of the chemotherapeutics cisplatin and vinblastine sulfate combined with a proprietary compound that facilitates penetration into the tumor tissue when injected directly into the tumor [294]. An earlier clinical trial demonstrated successful retention of the drugs within the tumor and morphological changes in the lesions with few side effects [295]. Previous animal models also demonstrated greater saturation of these drugs within the tumor compared with their normal intravenous delivery, and the compound also promoted an adaptive immune response that then successfully cleared noninjected metastases and immunized the animals against the cancer [293,294]. If successful, this innovative technology would simultaneously allow for targeted delivery of chemotherapeutics and generate an effective cancer vaccine where numerous previous attempts have failed. Nanoparticle-assisted delivery of drugs and visualization agents to hardto-target tumors are active areas of preclinical research, and excellent reviews are available on nanomedicine tumor penetration [296] and cellpenetrating peptides [297].
Conclusions Despite advances in the diagnosis and treatment of other cancers, there are still few options for identifying tumors of the pancreas although surgical resection (the only curative solution to PC) can be attempted [1,2]. There is promise as of late in characterizing serum markers that are more efficacious than CA19.9 in detecting not only early forms of PC but also in differentiating among common types of pancreatic lesions as well as other, similar diseases. However, the development of standardized marker panels has been slow, and at over 44,000 deaths per year [1], there is still a pressing need for innovation. The issue that has plagued therapeutic development for decades is the pancreatic tumor microenvironment, which is frequently ignored in the preclinical stage to the detriment of significant clinical progress. As a result, the next big discovery in PC may depend on conquering the stroma, whether through improved targeting of therapeutics via drug conjugation, the use of nanoparticles and penetration enhancers, or via stromal depletion. As always, it is still too early to know whether currently emerging technologies will be effective in the clinic. Indeed, any meaningful progress in reducing the burden of PC may require several more advances in our
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
351
understanding of the molecular pathology of PC. Nevertheless, if the number of innovative therapeutic and diagnostic strategies that have emerged are any indication, we may soon be one step closer to diminishing the burden of PC.
Acknowledgments This work was supported by the National Institute of Health Grant R01-DK81413 (to SAS) from the National Institute of Diabetes and Digestive and Kidney Diseases and by the UCF Reach for the Stars Award (to SAS). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute of Diabetes and Digestive and Kidney Diseases or the National Institutes of Health.
References [1] SEER cancer stat facts: pancreatic cancer. National Cancer Institute; 2019. https:// seer.cancer.gov/statfacts/html/pancreas.html. [2] Kim VM, Ahuja N. Early detection of pancreatic cancer. Chin J Cancer Res (Chungkuo yen cheng yen chiu) 2015;27(4):321e31. [3] Zhou B, Xu JW, Cheng YG, et al. Early detection of pancreatic cancer: where are we now and where are we going? Int J Cancer 2017;141(2):231e41. [4] Pancreatic cancer treatment. National Cancer Institute; 2019. https://www.cancer. gov/types/pancreatic/patient/pancreatic-treatment-pdq#section/all. [5] De La Cruz MS, Young AP, Ruffin MT. Diagnosis and management of pancreatic cancer. Am Fam Physician 2014;89(8):626e32. [6] Poruk KE, Firpo MA, Adler DG, Mulvihill SJ. Screening for pancreatic cancer. Ann Surg 2013;257(1):17e26. [7] Rossi ML, Rehman AA, Gondi CS. Therapeutic options for the management of pancreatic cancer. World J Gastroenterol 2014;20(32):11142e59. [8] Ilic M, Ilic I. Epidemiology of pancreatic cancer. World J Gastroenterol 2016;22(44):9694e705. [9] Zhang L, Sanagapalli S, Stoita A. Challenges in diagnosis of pancreatic cancer. World J Gastroenterol 2018;24(19):2047e60. [10] Pancreatic cancer - introduction. July 9, 2018. https://www.cancer.net/cancertypes/pancreatic-cancer/introduction. Cancer.Net. [11] Hansel DE, Kern SE, Hruban RH. Molecular pathogenesis of pancreatic cancer. Annu Rev Genom Hum Genet 2003;4:237e56. [12] Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010;467(7319):1114e7. [13] Distler M, Aust D, Weitz J, Pilarsky C, Grützmann R. Precursor lesions for sporadic pancreatic cancer: PanIN, IPMN, and MCN. BioMed Res Int 2014;2014:1e11. [14] Mettu NB, Abbruzzese JL. Clinical insights into the biology and treatment of pancreatic cancer. J Oncol Pract 2016;12(1):17e23. [15] Zamboni G, Hirabayashi K, Castelli P, Lennon AM. Precancerous lesions of the pancreas. Best Pract Res Clin Gastroenterol 2013;27(2):299e322. [16] Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet 2004;363(9414): 1049e57. [17] Maitra A, Hruban RH. Pancreatic cancer. Annu Rev Pathol 2008;3(1):157e88.
352
Theranostic Approach for Pancreatic Cancer
[18] Hruban RH, Maitra A, Goggins M. Update on pancreatic intraepithelial neoplasia. Int J Clin Exp Pathol 2008;1(4):306e16. [19] Norris AL, Roberts NJ, Jones S, et al. Familial and sporadic pancreatic cancer share the same molecular pathogenesis. Fam Cancer 2015;14(1):95e103. [20] Tomaszewska R, Okon K, Nowak K, Stachura J. HER-2/Neu expression as a progression marker in pancreatic intraepithelial neoplasia. Pol J Pathol 1998;49(2):83e92. [21] Chari ST, Kelly K, Hollingsworth MA, et al. Early detection of sporadic pancreatic cancer. Pancreas 2015;44(5):693e712. [22] Roberts NJ, Norris AL, Petersen GM, et al. Whole genome sequencing defines the genetic heterogeneity of familial pancreatic cancer. Cancer Discov 2016;6(2):166e75. [23] Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell 1988;53(4):549e54. [24] Morton JP, Timpson P, Karim SA, et al. Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc Natl Acad Sci USA 2010;107(1):246e51. [25] Bardeesy N, Cheng KH, Berger JH, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev 2006;20(22):3130e46. [26] Yachida S, White CM, Naito Y, et al. Clinical significance of the genetic landscape of pancreatic cancer and implications for identification of potential long-term survivors. Clin Cancer Res 2012;18(22):6339e47. [27] Whatcott CJ, Diep CH, Jiang P, et al. Desmoplasia in primary tumors and metastatic lesions of pancreatic cancer. Clin Cancer Res 2015;21(15):3561e8. [28] Whatcott CJ, Posner RG, Von Hoff DD, Han H. Desmoplasia and chemoresistance in pancreatic cancer. In: Grippo PJ, Munshi HG, editors. Pancreatic cancer and tumor microenvironment. Trivandrum, India: Transworld Research Network Transworld Research Network; 2012. [29] Xu Z, Pothula SP, Wilson JS, Apte MV. Pancreatic cancer and its stroma: a conspiracy theory. World J Gastroenterol 2014;20(32):11216e29. [30] Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res 2007;67(19):9518e27. [31] Nolan-Stevaux O, Lau J, Truitt ML, et al. GLI1 is regulated through smoothenedindependent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes Dev 2009;23(1):24e36. [32] Liyanage UK, Goedegebuure PS, Moore TT, et al. Increased prevalence of regulatory T cells (Treg) is induced by pancreas adenocarcinoma. J Immunother 2006;29(4):416e24. [33] Zhang Z, Ji S, Zhang B, et al. Role of angiogenesis in pancreatic cancer biology and therapy. Biomed Pharmacother 2018;108:1135e40. [34] King J, Bouvet M, Singh G, Williams J. Improving theranostics in pancreatic cancer. J Surg Oncol 2017;116(1):104e13. [35] Kannagi R. Carbohydrate antigen sialyl Lewis a–its pathophysiological significance and induction mechanism in cancer progression. Chang Gung Med J 2007;30(3):189e209. [36] Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet (Lond Engl) 2016;388(10039):73e85. [37] Lee KJ, Yi SW, Chung MJ, et al. Serum CA 19-9 and CEA levels as a prognostic factor in pancreatic adenocarcinoma. Yonsei Med J 2013;54(3):643.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
353
[38] Reitz D, Gerger A, Seidel J, et al. Combination of tumour markers CEA and CA19-9 improves the prognostic prediction in patients with pancreatic cancer. J Clin Pathol 2015;68(6):427e33. [39] Kaur S, Smith LM, Patel A, et al. A combination of MUC5AC and CA19-9 improves the diagnosis of pancreatic cancer: a multicenter study. Am J Gastroenterol 2017;112(1):172e83. [40] Sawada R, Sun SM, Wu X, et al. Human monoclonal antibodies to sialyl-Lewis (CA19.9) with potent CDC, ADCC, and antitumor activity. Clin Cancer Res 2011;17(5):1024e32. [41] Dimastromatteo J, Houghton JL, Lewis JS, Kelly KA. Challenges of pancreatic cancer. Cancer J (Sudbury Mass) 2015;21(3):188e93. [42] Rochefort MM, Girgis MD, Knowles SM, et al. A mutated anti-CA19-9 scFv-Fc for positron emission tomography of human pancreatic cancer xenografts. Mol Imaging Biol 2014;16(5):721e9. [43] Girgis MD, Federman N, Rochefort MM, et al. An engineered anti-CA19-9 cysdiabody for positron emission tomography imaging of pancreatic cancer and targeting of polymerized liposomal nanoparticles. J Surg Res 2013;185(1):45e55. [44] Girgis MD, Kenanova V, Olafsen T, McCabe KE, Wu AM, Tomlinson JS. AntiCA19-9 diabody as a PET imaging probe for pancreas cancer. J Surg Res 2011;170(2):169e78. [45] Houghton JL, Zeglis BM, Abdel-Atti D, et al. Site-specifically labeled CA19.9targeted immunoconjugates for the PET, NIRF, and multimodal PET/NIRF imaging of pancreatic cancer. Proc Natl Acad Sci USA 2015;112(52):15850e5. [46] Safety study of 90Y-hMN14 to treat pancreatic cancer. 2019. https://clinicaltrials. gov/ct2/show/NCT00041639?term¼labetuzumab&rank¼2. ClinicalTrials.gov. [47] Dotan E, Cohen SJ, Starodub AN, et al. Phase I/II trial of labetuzumab govitecan (anti-CEACAM5/SN-38 antibody-drug conjugate) in patients with refractory or relapsing metastatic colorectal cancer. J Clin Oncol 2017;35(29):3338e46. [48] Govindan SV, Cardillo TM, Moon SJ, Hansen HJ, Goldenberg DM. CEACAM5targeted therapy of human colonic and pancreatic cancer xenografts with potent labetuzumab-SN-38 immunoconjugates. Clin Cancer Res 2009;15(19):6052e61. [49] Dong W, Shi J, Yuan T, et al. Antibody-drug conjugates of 7-ethyl-10hydroxycamptothecin: sacituzumab govitecan and labetuzumab govitecan. Eur J Med Chem 2019;167:583e93. [50] Hajjar G, Sharkey RM, Burton J, et al. Phase I radioimmunotherapy trial with iodine-131–labeled humanized MN-14 anti-carcinoembryonic antigen monoclonal antibody in patients with metastatic gastrointestinal and colorectal cancer. Clin Colorectal Cancer 2002;2(1):31e42. [51] Sultana A, Shore S, Raraty MG, et al. Randomised phase I/II trial assessing the safety and efficacy of radiolabelled anti-carcinoembryonic antigen I(131) KAb201 antibodies given intra-arterially or intravenously in patients with unresectable pancreatic adenocarcinoma. BMC Cancer 2009;9:66. [52] Lwin TM, Miyake K, Murakami T, et al. Fluorescent humanized anti-CEA antibody specifically labels metastatic pancreatic cancer in a patient-derived orthotopic xenograft (PDOX) mouse model. Oncotarget 2018;9(99):37333e42. [53] Lwin TM, Murakami T, Miyake K, et al. Tumor-specific labeling of pancreatic cancer using a humanized anti-CEA antibody conjugated to a near-infrared fluorophore. Ann Surg Oncol 2018;25(4):1079e85. [54] DeLong JC, Murakami T, Yazaki PJ, Hoffman RM, Bouvet M. Near-infraredconjugated humanized anti-carcinoembryonic antigen antibody targets colon cancer in an orthotopic nude-mouse model. J Surg Res 2017;218:139e43.
354
Theranostic Approach for Pancreatic Cancer
[55] Yazaki PJ, Sherman MA, Shively JE, et al. Humanization of the anti-CEA T84.66 antibody based on crystal structure data. Protein Eng Des Sel 2004;17(5):481e9. [56] Immunoscintigraphy using radiolabeled monoclonal antibody in finding tumor cells in patients with colorectal cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ ct2/show/NCT00647153?term¼t84.66&rank¼1. [57] Wong JY, Chu DZ, Williams LE, et al. A phase I trial of (90)Y-DOTA-anti-CEA chimeric T84.66 (cT84.66) radioimmunotherapy in patients with metastatic CEAproducing malignancies. Cancer Biother Radiopharm 2006;21(2):88e100. [58] Wong JY, Shibata S, Williams LE, et al. A Phase I trial of 90Y-anti-carcinoembryonic antigen chimeric T84.66 radioimmunotherapy with 5-fluorouracil in patients with metastatic colorectal cancer. Clin Cancer Res 2003;9(16 Pt 1):5842e52. [59] Hassan R, Thomas A, Alewine C, Le DT, Jaffee EM, Pastan I. Mesothelin immunotherapy for cancer: ready for prime time? J Clin Oncol 2016;34(34):4171e9. [60] Baldo P, Cecco S. Amatuximab and novel agents targeting mesothelin for solid tumors. OncoTargets Ther 2017;10:5337e53. [61] An efficacy study of MORAb-009 in subjects with pancreatic cancer - study results. 2019. https://clinicaltrials.gov/ct2/show/results/NCT00570713?term¼amatuximab &rank¼7. ClinicalTrials.gov. [62] Anetumab ravtansine and atezolizumab in treating participants with advanced nonsmall cell lung cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/ NCT03455556?term¼anetumab%2Bravtansine&rank¼2. [63] Golfier S, Kopitz C, Kahnert A, et al. Anetumab ravtansine: a novel mesothelintargeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther 2014;13(6):1537e48. [64] A study of DMOT4039A in participants with unresectable pancreatic or platinumresistant ovarian cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/ show/NCT01469793. [65] Hassan R, Bullock S, Premkumar A, et al. Phase I study of SS1P, a recombinant antimesothelin immunotoxin given as a bolus I.V. infusion to patients with mesothelinexpressing mesothelioma, ovarian, and pancreatic cancers. Clin Cancer Res 2007;13(17):5144e9. [66] Hollevoet K, Mason-Osann E, Liu XF, Imhof-Jung S, Niederfellner G, Pastan I. In vitro and in vivo activity of the low-immunogenic antimesothelin immunotoxin RG7787 in pancreatic cancer. Mol Cancer Ther 2014;13(8):2040e9. [67] Le DT, Brockstedt DG, Nir-Paz R, et al. A live-attenuated Listeria vaccine (ANZ100) and a live-attenuated Listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: phase I studies of safety and immune induction. Clin Cancer Res 2012;18(3):858e68. [68] Safety & immunogenicity of JNJ-64041757, live-attenuated double-deleted Listeria immunotherapy, in subjects with non small cell lung cancer. 2019. https:// clinicaltrials.gov/ct2/show/NCT02592967. ClinicalTrials.gov. [69] CAR T cell receptor immunotherapy targeting mesothelin for patients with metastatic cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/ NCT01583686. [70] CART-meso in mesothelin expressing cancers. 2019. ClinicalTrials.gov. https:// clinicaltrials.gov/ct2/show/NCT02159716. [71] Malignant pleural disease treated with autologous T cells genetically engineered to target the cancer-cell surface antigen mesothelin. 2019. ClinicalTrials.gov. https:// clinicaltrials.gov/ct2/show/NCT02414269. [72] Morello A, Sadelain M, Adusumilli PS. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov 2016;6(2):133e46.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
355
[73] O’Hara M, Stashwick C, Haas AR, Tanyi JL. Mesothelin as a target for chimeric antigen receptor-modified T cells as anticancer therapy. Immunotherapy 2016;8(4):449e60. [74] Kalli M, Minia A, Pliaka V, Fotis C, Alexopoulos LG, Stylianopoulos T. Solid stressinduced migration is mediated by GDF15 through Akt pathway activation in pancreatic cancer cells. Sci Rep 2019;9(1):978. [75] Ratnam NM, Peterson JM, Talbert EE, et al. NF-kappaB regulates GDF-15 to suppress macrophage surveillance during early tumor development. J Clin Investig 2017;127(10):3796e809. [76] Zhou YF, Xu LX, Huang LY, et al. Combined detection of serum UL16-binding protein 2 and macrophage inhibitory cytokine-1 improves early diagnosis and prognostic prediction of pancreatic cancer. Oncol Lett 2014;8(5):2096e102. [77] Koopmann J, Rosenzweig CN, Zhang Z, et al. Serum markers in patients with resectable pancreatic adenocarcinoma: macrophage inhibitory cytokine 1 versus CA19-9. Clin Cancer Res 2006;12(2):442e6. [78] Wang X, Li Y, Tian H, et al. Macrophage inhibitory cytokine 1 (MIC-1/GDF15) as a novel diagnostic serum biomarker in pancreatic ductal adenocarcinoma. BMC Cancer 2014;14:578. [79] Jimenez-Luna C, Torres C, Ortiz R, et al. Proteomic biomarkers in body fluids associated with pancreatic cancer. Oncotarget 2018;9(23):16573e87. [80] Song J, Sokoll LJ, Pasay JJ, et al. Identification of serum biomarker panels for the early detection of pancreatic cancer. Cancer Epidemiol Biomarkers Prev 2019;28(1):174e82. [81] Hogendorf P, Durczynski A, Skulimowski A, Kumor A, Poznanska G, Strzelczyk J. Growth differentiation factor (GDF-15) concentration combined with Ca125 levels in serum is superior to commonly used cancer biomarkers in differentiation of pancreatic mass. Cancer Biomark 2018;21(3):505e11. [82] Mackman N. The role of tissue factor and factor VIIa in hemostasis. Anesth Analg 2009;108(5):1447e52. [83] Woei AJFJ, Tesselaar ME, Garcia Rodriguez P, Romijn FP, Bertina RM, Osanto S. Tissue factor-bearing microparticles and CA19.9: two players in pancreatic cancerassociated thrombosis? Br J Cancer 2016;115(3):332e8. [84] Kakkar AK, Lemoine NR, Scully MF, Tebbutt S, Williamson RC. Tissue factor expression correlates with histological grade in human pancreatic cancer. Br J Surg 1995;82(8):1101e4. [85] Hernandez R, England CG, Yang Y, et al. ImmunoPET imaging of tissue factor expression in pancreatic cancer with (89)Zr-Df-ALT-836. J Control Release 2017;264:160e8. [86] Nielsen CH, Jeppesen TE, Kristensen LK, et al. PET imaging of tissue factor in pancreatic cancer using 64Cu-labeled active site-inhibited factor VII. J Nucl Med 2016;57(7):1112e9. [87] Aung W, Tsuji AB, Sugyo A, et al. Near-infrared photoimmunotherapy of pancreatic cancer using an indocyanine green-labeled anti-tissue factor antibody. World J Gastroenterol 2018;24(48):5491e504. [88] Takashima H, Tsuji AB, Saga T, et al. Molecular imaging using an anti-human tissue factor monoclonal antibody in an orthotopic glioma xenograft model. Sci Rep 2017;7(1):12341. [89] Zhang Q, Wang H, Li H, et al. Chimeric antigen receptor-modified T Cells inhibit the growth and metastases of established tissue factor-positive tumors in NOG mice. Oncotarget 2017;8(6):9488e99.
356
Theranostic Approach for Pancreatic Cancer
[90] Breij EC, de Goeij BE, Verploegen S, et al. An antibody-drug conjugate that targets tissue factor exhibits potent therapeutic activity against a broad range of solid tumors. Cancer Res 2014;74(4):1214e26. [91] Dong H, Qian D, Wang Y, et al. Survivin expression and serum levels in pancreatic cancer. World J Surg Oncol 2015;13:189. [92] Santarelli A, Mascitti M, Lo Russo L, et al. Survivin-based treatment strategies for squamous cell carcinoma. Int J Mol Sci 2018;19(4). [93] Talbot DC, Ranson M, Davies J, et al. Tumor survivin is downregulated by the antisense oligonucleotide LY2181308: a proof-of-concept, first-in-human dose study. Clin Cancer Res 2010;16(24):6150e8. [94] Tanioka M, Nokihara H, Yamamoto N, et al. Phase I study of LY2181308, an antisense oligonucleotide against survivin, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2011;68(2):505e11. [95] A phase I/II study of paclitaxel, carboplatin and YM155 (survivin suppressor) in subjects with solid tumors (phase I) and advanced non-small cell lung carcinoma (phase II) - study results. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/ results/NCT01100931?term¼ym155&draw¼2&rank¼7. [96] Tanaka T, Kitamura H, Inoue R, et al. Potential survival benefit of anti-apoptosis protein: survivin-derived peptide vaccine with and without interferon alpha therapy for patients with advanced or recurrent urothelial cancer–results from phase I clinical trials. Clin Dev Immunol 2013;2013:262967. [97] A feasibility and safety study of vaccination with poly-ICLC and peptide-pulsed dendritic cells in patients with metastatic, locally advanced, unresectable, or recurrent pancreatic adenocarcinoma. 2019. https://clinicaltrials.gov/ct2/show/ NCT01410968?term¼survivin&cond¼pancreatic%2Bcancer&rank¼3. ClinicalTrials.gov. [98] TAA specific cytotoxic T lymphocytes in patients with pancreatic cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03192462?term¼survivin& cond¼pancreatic%2Bcancer&rank¼2. [99] Sheer DG, Schlom J, Cooper HL. Purification and composition of the human tumorassociated glycoprotein (TAG-72) defined by monoclonal antibodies CC49 and B72.3. Cancer Res 1988;48(23):6811e8. [100] Louhimo J, Alfthan H, Stenman UH, Haglund C. Serum HCG beta and CA 72-4 are stronger prognostic factors than CEA, CA 19-9 and CA 242 in pancreatic cancer. Oncology 2004;66(2):126e31. [101] Bohdiewicz PJ. Indium-111 satumomab pendetide: the first FDA-approved monoclonal antibody for tumor imaging. J Nucl Med Technol 1998;26(3):155e63. Quiz 170-151. [102] Cho J, Kim KM, Kim HC, et al. The prognostic role of tumor associated glycoprotein 72 (TAG-72) in stage II and III colorectal adenocarcinoma. Pathol Res Pract 2019;215(1):171e6. [103] Gong L, Ding H, Long NE, et al. A 3E8.scFv.Cys-IR800 conjugate targeting TAG72 in an orthotopic colorectal cancer model. Mol Imaging Biol 2018;20(1):47e54. [104] Yoon SO, Lee TS, Kim SJ, et al. Construction, affinity maturation, and biological characterization of an anti-tumor-associated glycoprotein-72 humanized antibody. J Biol Chem 2006;281(11):6985e92. [105] Ding H, Carlton MM, Povoski SP, et al. Site specific discrete PEGylation of (124)Ilabeled mCC49 Fab0 fragments improves tumor MicroPET/CT imaging in mice. Bioconjug Chem 2013;24(11):1945e54. [106] Murad JP, Kozlowska AK, Lee HJ, et al. Effective targeting of TAG72(þ) peritoneal ovarian tumors via regional delivery of CAR-engineered T cells. Front Immunol 2018;9:2268.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
357
[107] Siddiqui AA, Jones E, Andrade D, et al. Osteopontin splice variant as a potential marker for metastatic disease in pancreatic adenocarcinoma. J Gastroenterol Hepatol 2014;29(6):1321e7. [108] Zhao H, Chen Q, Alam A, et al. The role of osteopontin in the progression of solid organ tumour. Cell Death Dis 2018;9(3):356. [109] Lazar M, Sullivan J, Chipitsyna G, et al. Involvement of osteopontin in the matrixdegrading and proangiogenic changes mediated by nicotine in pancreatic cancer cells. J Gastrointest Surg 2010;14(10):1566e77. [110] Poruk KE, Firpo MA, Scaife CL, et al. Serum osteopontin and tissue inhibitor of metalloproteinase 1 as diagnostic and prognostic biomarkers for pancreatic adenocarcinoma. Pancreas 2013;42(2):193e7. [111] Dai J, Li B, Shi J, et al. A humanized anti-osteopontin antibody inhibits breast cancer growth and metastasis in vivo. Cancer Immunol Immunother 2010;59(3):355e66. [112] Kou G, Shi J, Chen L, et al. A bispecific antibody effectively inhibits tumor growth and metastasis by simultaneous blocking vascular endothelial growth factor A and osteopontin. Cancer Lett 2010;299(2):130e6. [113] Study of intratumoral hypoxia using pre-operative administration of pimonidazole. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01248637?term¼ osteopontin&cond¼pancreatic%2Bcancer&rank¼1. [114] Feng L, Qi Q, Wang P, et al. Serum levels of IL-6, IL-8, and IL-10 are indicators of prognosis in pancreatic cancer. J Int Med Res 2018;46(12):5228e36. [115] Chen Y, Shi M, Yu GZ, et al. Interleukin-8, a promising predictor for prognosis of pancreatic cancer. World J Gastroenterol 2012;18(10):1123e9. [116] Ebrahimi B, Tucker SL, Li D, Abbruzzese JL, Kurzrock R. Cytokines in pancreatic carcinoma: correlation with phenotypic characteristics and prognosis. Cancer 2004;101(12):2727e36. [117] Mitsunaga S, Ikeda M, Shimizu S, et al. Serum levels of IL-6 and IL-1beta can predict the efficacy of gemcitabine in patients with advanced pancreatic cancer. Br J Cancer 2013;108(10):2063e9. [118] Zhang Y, Yan W, Collins MA, et al. Interleukin-6 is required for pancreatic cancer progression by promoting MAPK signaling activation and oxidative stress resistance. Cancer Res 2013;73(20):6359e74. [119] Chen L, Fan J, Chen H, et al. The IL-8/CXCR1 axis is associated with cancer stem cell-like properties and correlates with clinical prognosis in human pancreatic cancer cases. Sci Rep 2014;4:5911. [120] Dennis KL, Blatner NR, Gounari F, Khazaie K. Current status of interleukin-10 and regulatory T-cells in cancer. Curr Opin Oncol 2013;25(6):637e45. [121] Sato T, Terai M, Tamura Y, Alexeev V, Mastrangelo MJ, Selvan SR. Interleukin 10 in the tumor microenvironment: a target for anticancer immunotherapy. Immunol Res 2011;51(2e3):170e82. [122] Yao X, Huang J, Zhong H, et al. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol Ther 2014;141(2):125e39. [123] Angevin E, Tabernero J, Elez E, et al. A phase I/II, multiple-dose, dose-escalation study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res 2014;20(8):2192e204. [124] Study of nab-paclitaxel and gemcitabine with or without tocilizumab in pancreatic cancer patients. 2019. https://clinicaltrials.gov/ct2/show/NCT02767557. Clinical Trials.gov. [125] Zhang L, Luan B, Adler A, Eichten A, Daly C, Thurston G. Abstract 2723: Sarilumab (REGN88), a fully-human anti-IL6R antibody, inhibits tumor growth in preclinical models, as a single agent and in combination with the VEGF blocker aflibercept. Cancer Res 2012;72(8 Suppl.):2723.
358
Theranostic Approach for Pancreatic Cancer
[126] Bayliss TJ, Smith JT, Schuster M, Dragnev KH, Rigas JR. A humanized anti-IL-6 antibody (ALD518) in non-small cell lung cancer. Exp Opin Biol Ther 2011;11(12):1663e8. [127] Rose-John S. The soluble interleukin 6 receptor: advanced therapeutic options in inflammation. Clin Pharmacol Ther 2017;102(4):591e8. [128] Kim GW, Lee NR, Pi RH, et al. IL-6 inhibitors for treatment of rheumatoid arthritis: past, present, and future. Arch Pharm Res 2015;38(5):575e84. [129] Oji Y, Nakamori S, Fujikawa M, et al. Overexpression of the Wilms’ tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci 2004;95(7):583e7. [130] Cheever MA, Allison JP, Ferris AS, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res 2009;15(17):5323e37. [131] Kajihara M, Takakura K, Kanai T, et al. Advances in inducing adaptive immunity using cell-based cancer vaccines: clinical applications in pancreatic cancer. World J Gastroenterol 2016;22(18):4446e58. [132] Dendritic cell immunotherapy for the patients with pancreatic cancer. 2019. https://clinicaltrials.gov/ct2/show/NCT03114631?term¼wt1&cond¼Pancreatic% 2BCancer&rank¼1. ClinicalTrials.gov. [133] Zhang W, Lu X, Cui P, et al. Phase I/II clinical trial of a Wilms’ tumor 1-targeted dendritic cell vaccination-based immunotherapy in patients with advanced cancer. Cancer Immunol Immunother 2019;68(1):121e30. [134] Akahori Y, Wang L, Yoneyama M, et al. Antitumor activity of CAR-T cells targeting the intracellular oncoprotein WT1 can be enhanced by vaccination. Blood 2018;132(11):1134e45. [135] Rafiq S, Purdon TJ, Daniyan AF, et al. Optimized T-cell receptor-mimic chimeric antigen receptor T cells directed toward the intracellular Wilms Tumor 1 antigen. Leukemia 2017;31(8):1788e97. [136] Nagaraju GP, Dontula R, El-Rayes BF, Lakka SS. Molecular mechanisms underlying the divergent roles of SPARC in human carcinogenesis. Carcinogenesis 2014;35(5):967e73. [137] Tanaka HY, Kitahara K, Sasaki N, et al. Pancreatic stellate cells derived from human pancreatic cancer demonstrate aberrant SPARC-dependent ECM remodeling in 3D engineered fibrotic tissue of clinically relevant thickness. Biomaterials 2019;192:355e67. [138] Desai NP, Trieu V, Hwang LY, Wu R, Soon-Shiong P, Gradishar WJ. Improved effectiveness of nanoparticle albumin-bound (nab) paclitaxel versus polysorbate-based docetaxel in multiple xenografts as a function of HER2 and SPARC status. Anti Cancer Drugs 2008;19(9):899e909. [139] Desai N, Trieu V, Damascelli B, Soon-Shiong P. SPARC expression correlates with tumor response to albumin-bound paclitaxel in head and neck cancer patients. Transl Oncol 2009;2(2):59e64. [140] Martín AJ, Alfonso PG, Rupérez AB, Jiménez MM. Nab-paclitaxel plus gemcitabine as first-line palliative chemotherapy in a patient with metastatic pancreatic cancer with Eastern Cooperative Oncology Group performance status of 2. Oncol Lett 2016;12(1):727e30. [141] Kim B, Lee C, Lee ES, Shin BS, Youn YS. Paclitaxel and curcumin co-bound albumin nanoparticles having antitumor potential to pancreatic cancer. Asian J Pharm Sci 2016;11(6):708e14. [142] Alemi A, Zavar Reza J, Haghiralsadat F, et al. Paclitaxel and curcumin coadministration in novel cationic PEGylated niosomal formulations exhibit enhanced synergistic antitumor efficacy. J Nanobiotechnol 2018;16(1):28.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
359
[143] Wei Y, Pu X, Zhao L. Preclinical studies for the combination of paclitaxel and curcumin in cancer therapy (review). Oncol Rep 2017;37(6):3159e66. [144] Von Hoff DD, Ramanathan RK, Borad MJ, et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol 2011;29(34):4548e54. [145] Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369(18):1691e703. [146] Yang J, Lv Q, Wei W, et al. Bioresponsive albumin-conjugated paclitaxel prodrugs for cancer therapy. Drug Deliv 2018;25(1):807e14. [147] Paclitaxel protein bound plus cisplatin plus gemcitabine and paricalcitol for pancreatic adenocarcinoma (NABPLAGEMD). 2019. ClinicalTrials.gov. https:// clinicaltrials.gov/ct2/show/NCT03415854?term¼hyaluronic%2Bacid&cond¼ pancreatic%2Bcancer&rank¼6. [148] Zheng L. PD-L1 expression in pancreatic cancer. J Natl Cancer Inst 2017;109(6). [149] Torphy RJ, Zhu Y, Schulick RD. Immunotherapy for pancreatic cancer: barriers and breakthroughs. Ann Gastroenterol Surg 2018;2(4):274e81. [150] Cheng X, Zhao G, Zhao Y. Combination immunotherapy approaches for pancreatic cancer treatment. Can J Gastroenterol Hepatol 2018;2018:6240467. [151] Blando J, Sharma A, Higa MG, et al. Comparison of immune infiltrates in melanoma and pancreatic cancer highlights VISTA as a potential target in pancreatic cancer. Proc Natl Acad Sci USA 2019;116(5):1692e7. [152] Lines JL, Pantazi E, Mak J, et al. VISTA is an immune checkpoint molecule for human T cells. Cancer Res 2014;74(7):1924e32. [153] A study of safety, pharmacokinetics, pharmacodynamics of JNJ-61610588 in participants with advanced cancer. 2019. https://clinicaltrials.gov/ct2/show/ NCT02671955. ClinicalTrials.gov. [154] Sato N, Kohi S, Hirata K, Goggins M. Role of hyaluronan in pancreatic cancer biology and therapy: once again in the spotlight. Cancer Sci 2016;107(5):569e75. [155] Shepard HM. Breaching the castle walls: hyaluronan depletion as a therapeutic approach to cancer therapy. Front Oncol 2015;5:192. [156] Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013;62(1):112e20. [157] Zoltan-Jones A, Huang L, Ghatak S, Toole BP. Elevated hyaluronan production induces mesenchymal and transformed properties in epithelial cells. J Biol Chem 2003;278(46):45801e10. [158] Thompson CB, Shepard HM, O’Connor PM, et al. Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Mol Cancer Ther 2010;9(11):3052e64. [159] Second-line study of PEGPH20 and pembro for HA high metastatic PDAC. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03634332? term¼hyaluronic%2Bacid&cond¼pancreatic%2Bcancer&rank¼1. [160] Study of gemcitabine, nab-paclitaxel, PEGPH20 and rivaroxaban for advanced pancreatic adenocarcinoma. 2019. https://clinicaltrials.gov/ct2/show/NCT02921022? term¼hyaluronic%2Bacid&cond¼pancreatic%2Bcancer&rank¼2. ClinicalTrials.gov. [161] A trial of PEGPH20 in combination with avelumab in chemotherapy resistant pancreatic cancer. 2019. https://clinicaltrials.gov/ct2/show/NCT03481920?term¼ hyaluronic%2Bacid&cond¼pancreatic%2Bcancer&rank¼3. ClinicalTrials.gov. [162] Study of gemcitabine þ PEGPH20 vs gemcitabine alone in stage IV previously untreated pancreatic cancer. 2019. https://clinicaltrials.gov/ct2/show/NCT01453153? term¼hyaluronic%2Bacid&cond¼pancreatic%2Bcancer&rank¼4. ClinicalTrials.gov.
360
Theranostic Approach for Pancreatic Cancer
[163] PEGPH20 plus gemcitabine with radiotherapy in patients with localized, unresectable pancreatic cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/ NCT02910882?term¼hyaluronic%2Bacid&cond¼pancreatic%2Bcancer&rank¼5. [164] S1313, PEGPH20 in treating patients with newly diagnosed metastatic pancreatic cancer. 2019. https://clinicaltrials.gov/ct2/show/NCT01959139?term¼hyaluronic%2Bacid& cond¼pancreatic%2Bcancer&rank¼7. ClinicalTrials.gov. [165] A study of PEGylated recombinant human hyaluronidase in combination with nabpaclitaxel plus gemcitabine compared with placebo plus nab-paclitaxel and gemcitabine in participants with hyaluronan-high stage IV previously untreated pancreatic ductal adenocarcinoma. 2019. https://clinicaltrials.gov/ct2/show/NCT02715804. ClinicalTrials.gov. [166] Infante JR, Korn RL, Rosen LS, et al. Phase 1 trials of PEGylated recombinant human hyaluronidase PH20 in patients with advanced solid tumours. Br J Cancer 2018;118(2):153e61. [167] Ramanathan RK, McDonough SL, Philip PA, et al. Phase IB/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. J Clin Oncol 2019. Jco1801295. [168] Hingorani SR, Harris WP, Beck JT, et al. Phase ib study of PEGylated recombinant human hyaluronidase and gemcitabine in patients with advanced pancreatic cancer. Clin Cancer Res 2016;22(12):2848e54. [169] Bhatia R, Gautam SK, Cannon A, et al. Cancer-associated mucins: role in immune modulation and metastasis. Cancer Metastasis Rev 2019. https://doi.org/10.1007/ s10555-018-09775-0. [170] Torres MP, Chakraborty S, Souchek J, Batra SK. Mucin-based targeted pancreatic cancer therapy. Curr Pharm Des 2012;18(17):2472e81. [171] Chaturvedi P, Singh AP, Moniaux N, et al. MUC4 mucin potentiates pancreatic tumor cell proliferation, survival, and invasive properties and interferes with its interaction to extracellular matrix proteins. Mol Cancer Res 2007;5(4):309e20. [172] Kohlgraf KG, Gawron AJ, Higashi M, et al. Contribution of the MUC1 tandem repeat and cytoplasmic tail to invasive and metastatic properties of a pancreatic cancer cell line. Cancer Res 2003;63(16):5011e20. [173] Singh AP, Moniaux N, Chauhan SC, Meza JL, Batra SK. Inhibition of MUC4 expression suppresses pancreatic tumor cell growth and metastasis. Cancer Res 2004;64(2):622e30. [174] Tsutsumida H, Swanson BJ, Singh PK, et al. RNA interference suppression of MUC1 reduces the growth rate and metastatic phenotype of human pancreatic cancer cells. Clin Cancer Res 2006;12(10):2976e87. [175] Jahan R, Macha MA, Rachagani S, et al. Axed MUC4 (MUC4/X) aggravates pancreatic malignant phenotype by activating integrin-beta1/FAK/ERK pathway. Biochim Biophys Acta Mol Basis Dis 2018;1864(8):2538e49. [176] Price MR, Rye PD, Petrakou E, et al. Summary report on the ISOBM TD-4 Workshop: analysis of 56 monoclonal antibodies against the MUC1 mucin. San Diego, Calif., November 17e23, 1996. Tumour Biol 1998;19(Suppl. 1):1e20. [177] Tarp MA, Clausen H. Mucin-type O-glycosylation and its potential use in drug and vaccine development. Biochim Biophys Acta 2008;1780(3):546e63. [178] Ohyabu N, Hinou H, Matsushita T, et al. An essential epitope of anti-MUC1 monoclonal antibody KL-6 revealed by focused glycopeptide library. J Am Chem Soc 2009;131(47):17102e9. [179] Naito S, Takahashi T, Onoda J, et al. Generation of novel anti-MUC1 monoclonal antibodies with designed carbohydrate specificities using MUC1 glycopeptide library. ACS Omega 2017;2(11):7493e505.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
361
[180] Wu G, Maharjan S, Kim D, et al. A novel monoclonal antibody targets Mucin1 and attenuates growth in pancreatic cancer model. Int J Mol Sci 2018;19(7). [181] Helft PR, Schilsky RL, Hoke FJ, et al. A phase I study of cantuzumab mertansine administered as a single intravenous infusion once weekly in patients with advanced solid tumors. Clin Cancer Res 2004;10(13):4363e8. [182] Danielczyk A, Stahn R, Faulstich D, et al. PankoMab: a potent new generation anti-tumour MUC1 antibody. Cancer Immunol Immunother 2006;55(11): 1337e47. [183] PankoMab-GEXÔ versus Placebo as maintenance therapy in advanced ovarian cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01899599? term¼pankomab&rank¼2. [184] Fiedler W, DeDosso S, Cresta S, et al. A phase I study of PankoMab-GEX, a humanised glyco-optimised monoclonal antibody to a novel tumour-specific MUC1 glycopeptide epitope in patients with advanced carcinomas. Eur J Cancer (Oxf Engl 1990) 2016;63:55e63. [185] Han S, Jin G, Wang L, et al. The role of PAM4 in the management of pancreatic cancer: diagnosis, radioimmunodetection, and radioimmunotherapy. J Immunol Res 2014;2014:268479. [186] Liu D, Chang CH, Gold DV, Goldenberg DM. Identification of PAM4 (clivatuzumab)-reactive epitope on MUC5AC: a promising biomarker and therapeutic target for pancreatic cancer. Oncotarget 2015;6(6):4274e85. [187] Picozzi VJ, Ramanathan RK, Lowery MA, et al. (90)Y-clivatuzumab tetraxetan with or without low-dose gemcitabine: a phase Ib study in patients with metastatic pancreatic cancer after two or more prior therapies. Eur J Cancer (Oxf Engl 1990) 2015;51(14):1857e64. [188] Gautam SK, Kumar S, Cannon A, et al. MUC4 mucin- a therapeutic target for pancreatic ductal adenocarcinoma. Exp Opin Ther Targets 2017;21(7):657e69. [189] Jain M, Venkatraman G, Moniaux N, et al. Monoclonal antibodies recognizing the non-tandem repeat regions of the human mucin MUC4 in pancreatic cancer. PLoS One 2011;6(8):e23344. [190] Moniaux N, Varshney GC, Chauhan SC, et al. Generation and characterization of anti-MUC4 monoclonal antibodies reactive with normal and cancer cells in humans. J Histochem Cytochem 2004;52(2):253e61. [191] Wang G, Lipert RJ, Jain M, et al. Detection of the potential pancreatic cancer marker MUC4 in serum using surface-enhanced Raman scattering. Anal Chem 2011;83(7):2554e61. [192] Wei J, Gao W, Wu J, et al. Dendritic cells expressing a combined PADRE/MUC4derived polyepitope DNA vaccine induce multiple cytotoxic T-cell responses. Cancer Biother Radiopharm 2008;23(1):121e8. [193] Wu J, Wei J, Meng K, et al. Identification of an HLA-A*0201-restrictive CTL epitope from MUC4 for applicable vaccine therapy. Immunopharmacol Immunotoxicol 2009;31(3):468e76. [194] Vaccination of prostate cancer patients with MUC-2-KLH conjugate plus the immunological adjuvant QS21. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ ct2/show/NCT00698711?term¼muc2&rank¼1. [195] Vaccine therapy plus QS21 in treating patients with progressive prostate cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT00004929?term¼muc2& rank¼2. [196] Vaccine therapy plus QS21 in treating patients with prostate cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT00036933?term¼muc2& rank¼3.
362
Theranostic Approach for Pancreatic Cancer
[197] Vaccine therapy plus biological therapy in treating patients with prostate cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT00016146?term¼muc2& rank¼4. [198] Beatty GL, Chiorean EG, Fishman MP, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science (New York NY) 2011;331(6024):1612e6. [199] Vonderheide RH, Bajor DL, Winograd R, Evans RA, Bayne LJ, Beatty GL. CD40 immunotherapy for pancreatic cancer. Cancer Immunol Immunother 2013;62(5):949e54. [200] Vonderheide RH, Glennie MJ. Agonistic CD40 antibodies and cancer therapy. Clin Cancer Res 2013;19(5):1035e43. [201] Beatty GL, Torigian DA, Chiorean EG, et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res 2013;19(22):6286e95. [202] Vonderheide RH, Burg JM, Mick R, et al. Phase I study of the CD40 agonist antibody CP-870,893 combined with carboplatin and paclitaxel in patients with advanced solid tumors. OncoImmunology 2013;2(1):e23033. [203] A phase I study of the chimeric anti-CD40 monoclonal antibody ChiLob 7/4 to treat advanced malignancies refractory to conventional anti-cancer treatment. 2019. https://clinicaltrials.gov/ct2/show/NCT01561911. ClinicalTrials.gov. [204] Chou A, Waddell N, Cowley MJ, et al. Clinical and molecular characterization of HER2 amplified-pancreatic cancer. Genome Med 2013;5(8):78. [205] Harder J, Ihorst G, Heinemann V, et al. Multicentre phase II trial of trastuzumab and capecitabine in patients with HER2 overexpressing metastatic pancreatic cancer. Br J Cancer 2012;106(6):1033e8. [206] Yamanaka Y, Friess H, Kobrin MS, et al. Overexpression of HER2/neu oncogene in human pancreatic carcinoma. Hum Pathol 1993;24(10):1127e34. [207] Mangray S, King TC. Molecular pathobiology of pancreatic adenocarcinoma. Front Biosci 1998;3:D1148e60. [208] Karanikas M, Esempidis A, Chasan ZTM, et al. Pancreatic cancer from molecular pathways to treatment. Opinion 2016;7(10):1328e39. [209] Liu H, Sun M, Liu Z, et al. KRAS-enhanced macropinocytosis and reduced FcRnmediated recycling sensitize pancreatic cancer to albumin-conjugated drugs. J Control Release 2019;296:40e53. [210] Yao W, Rose JL, Wang W, et al. Syndecan 1 is a critical mediator of macropinocytosis in pancreatic cancer. Nature 2019;568(7752):410e4. [211] Wöll S, Schlitter AM, Dhaene K, et al. Claudin 18.2 is a target for IMAB362 antibody in pancreatic neoplasms. Int J Cancer 2014;134(3):731e9. [212] Singh P, Toom S, Huang Y. Anti-claudin 18.2 antibody as new targeted therapy for advanced gastric cancer. J Hematol Oncol 2017;10(1). [213] Tureci, Mitnacht-Kraus R, Woll S, Yamada T, Sahin U. Characterization of zolbetuximab in pancreatic cancer models. OncoImmunology 2019;8(1):e1523096. [214] A study of zolbetuximab (IMAB362) plus CAPOX compared with placebo plus CAPOX as first-line treatment of subjects with claudin (CLDN) 18.2-positive, HER2-negative, locally advanced unresectable or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma. 2019. ClinicalTrials.gov. https://clinicaltrials. gov/ct2/show/NCT03653507?term¼claudin%2B18.2&rank¼8. [215] A study to assess the antitumor activity, safety, pharmacokinetics and biomarkers of zolbetuximab (IMAB362) in participants with claudin (CLDN) 18.2 positive, metastatic or advanced unresectable gastric and gastroesophageal junction (GEJ) adenocarcinoma. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/ NCT03505320?term¼claudin%2B18.2&rank¼7.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
363
[216] A phase 3 efficacy, safety and tolerability study of zolbetuximab (experimental drug) plus mFOLFOX6 chemotherapy compared to placebo plus mFOLFOX6 as treatment for gastric and gastroesophageal junction (GEJ) cancer. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03504397?term¼claudin%2B18.2&rank¼5. [217] A study to assess the antitumor activity and safety of IMAB362 in combination with nab-paclitaxel and gemcitabine (Nab-P þ GEM) as first line treatment in subjects with claudin 18.2 (CLDN18.2) positive, metastatic pancreatic adenocarcinoma. 2019. https://clinicaltrials.gov/ct2/show/NCT03816163?term¼claudin%2B18.2&rank¼3. ClinicalTrials.gov. [218] Clinical study of CAR-CLD18 T cells in patients with advanced gastric adenocarcinoma and pancreatic adenocarcinoma. 2019. https://clinicaltrials.gov/ct2/show/ NCT03159819?term¼claudin%2B18.2&rank¼2. ClinicalTrials.gov. [219] Chimeric antigen receptor T cells targeting claudin18.2 in solid tumors. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03874897?term¼ claudin%2B18.2&rank¼1. [220] A phase 1, open-label study of LCAR-C182A/LCAR-C182B/LCAR-C182C cells, a chimeric antigen receptor T-cell (CAR-T). 2019. ClinicalTrials.gov. https:// clinicaltrials.gov/ct2/show/NCT03890198?term¼claudinþ18.2&rank¼4. [221] Clinical study of redirected autologous T cells with a chimeric antigen receptor in patients with malignant tumors. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ ct2/show/NCT03302403?term¼claudin%2B18.2&rank¼6. [222] Di Carlo C, Brandi J, Cecconi D. Pancreatic cancer stem cells: perspectives on potential therapeutic approaches of pancreatic ductal adenocarcinoma. World J Stem Cells 2018;10(11):172e82. [223] Fitzgerald TL, McCubrey JA. Pancreatic cancer stem cells: association with cell surface markers, prognosis, resistance, metastasis and treatment. Adv Biol Regul 2014;56:45e50. [224] Valle S, Martin-Hijano L, Alcalá S, Alonso-Nocelo M, Sainz JrJr. The ever-evolving concept of the cancer stem cell in pancreatic cancer. Cancers (Basel) 2018;10(2). [225] Hermann PC, Huber SL, Herrler T, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 2007;1(3):313e23. [226] Li C, Heidt DG, Dalerba P, et al. Identification of pancreatic cancer stem cells. Cancer Res 2007;67(3):1030e7. [227] Jia Y, Gu D, Wan J, et al. The role of GLI-SOX2 signaling axis for gemcitabine resistance in pancreatic cancer. Oncogene 2019;38(10):1764e77. [228] Zhang Z, Duan Q, Zhao H, et al. Gemcitabine treatment promotes pancreatic cancer stemness through the Nox/ROS/NF-kappaB/STAT3 signaling cascade. Cancer Lett 2016;382(1):53e63. [229] Zhang Z, Han H, Rong Y, et al. Hypoxia potentiates gemcitabine-induced stemness in pancreatic cancer cells through AKT/Notch1 signaling. J Exp Clin Cancer Res 2018;37(1):291. [230] Smigiel JM, Parameswaran N, Jackson MW. Targeting pancreatic cancer cell plasticity: the latest in therapeutics. Cancers (Basel) 2018;10(1). [231] Krishnamurthy N, Kurzrock R. Targeting the Wnt/beta-catenin pathway in cancer: update on effectors and inhibitors. Cancer Treat Rev 2018;62:50e60. [232] Li QR, Zhao H, Zhang XS, Lang H, Yu K. Novel-smoothened inhibitors for therapeutic targeting of naive and drug-resistant hedgehog pathway-driven cancers. Acta Pharmacol Sin 2019;40(2):257e67. [233] Rimkus T, Carpenter R, Qasem S, Chan M, Lo H-W. Targeting the sonic hedgehog signaling pathway: review of smoothened and GLI inhibitors. Cancers 2016;8(2):22.
364
Theranostic Approach for Pancreatic Cancer
[234] Zhang X, Hao J. Development of anticancer agents targeting the Wnt/b-catenin signaling. Am J Cancer Res 2015;5(8):2344e60. [235] Varillas JI, Zhang J, Chen K, et al. Microfluidic isolation of circulating tumor cells and cancer stem-like cells from patients with pancreatic ductal adenocarcinoma. Theranostics 2019;9(5):1417e25. [236] Kim WT, Ryu CJ. Cancer stem cell surface markers on normal stem cells. BMB Rep 2017;50(6):285e98. [237] Chen C, Zhao S, Karnad A, Freeman JW. The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol 2018;11(1):64. [238] Luo Y, Prestwich GD. Synthesis and selective cytotoxicity of a hyaluronic acidantitumor bioconjugate. Bioconjug Chem 1999;10(5):755e63. [239] Serafino A, Zonfrillo M, Andreola F, et al. CD44-targeting for antitumor drug delivery: a new SN-38-hyaluronan bioconjugate for locoregional treatment of peritoneal carcinomatosis. Curr Cancer Drug Targets 2011;11(5):572e85. [240] Journo-Gershfeld G, Kapp D, Shamay Y, Kopecek J, David A. Hyaluronan oligomers-HPMA copolymer conjugates for targeting paclitaxel to CD44overexpressing ovarian carcinoma. Pharmaceut Res 2012;29(4):1121e33. [241] Ren Y, Zhao T, Mao G, et al. Antitumor activity of hyaluronic acid-selenium nanoparticles in Heps tumor mice models. Int J Biol Macromol 2013;57:57e62. [242] El-Dakdouki MH, Xia J, Zhu DC, et al. Assessing the in vivo efficacy of doxorubicin loaded hyaluronan nanoparticles. ACS Appl Mater Interfaces 2014;6(1):697e705. [243] Huang WY, Lin JN, Hsieh JT, et al. Nanoparticle targeting CD44-positive cancer cells for site-specific drug delivery in prostate cancer therapy. ACS Appl Mater Interfaces 2016;8(45):30722e34. [244] Lee JY, Chung SJ, Cho HJ, Kim DD. Iodinated hyaluronic acid oligomer-based nanoassemblies for tumor-targeted drug delivery and cancer imaging. Biomaterials 2016;85:218e31. [245] Dubey RD, Klippstein R, Wang JT, et al. Novel hyaluronic acid conjugates for dual nuclear imaging and therapy in CD44-expressing tumors in mice in vivo. Nanotheranostics 2017;1(1):59e79. [246] Han NK, Shin DH, Kim JS, Weon KY, Jang CY, Kim JS. Hyaluronan-conjugated liposomes encapsulating gemcitabine for breast cancer stem cells. Int J Nanomed 2016;11:1413e25. [247] Riker A, Libutti SK, Bartlett DL. Advances in the early detection, diagnosis, and staging of pancreatic cancer. Surg Oncol 1997;6(3):157e69. [248] Granata V, Fusco R, Catalano O, et al. Multidetector computer tomography in the pancreatic adenocarcinoma assessment: an update. Infect Agents Cancer 2016;11(1). [249] Luz LP, Al-Haddad MA, Sey MS, DeWitt JM. Applications of endoscopic ultrasound in pancreatic cancer. World J Gastroenterol 2014;20(24):7808e18. [250] Adamska A, Domenichini A, Falasca M. Pancreatic ductal adenocarcinoma: current and evolving therapies. Int J Mol Sci 2017;18(7):1338. [251] Ansari NA, Ramalho M, Semelka RC, Buonocore V, Gigli S, Maccioni F. Role of magnetic resonance imaging in the detection and characterization of solid pancreatic nodules: an update. World J Radiol 2015;7(11):361e74. [252] Serrano OK, Chaudhry MA, Leach SD. The role of PET scanning in pancreatic cancer. Adv Surg 2010;44(1):313e25. [253] Scialpi M, Reginelli A, D’Andrea A, et al. Pancreatic tumors imaging: an update. Int J Surg 2016;28:S142e55. [254] Willett CG, Daly WJ, Warshaw AL. CA 19-9 is an index of response to neoadjunctive chemoradiation therapy in pancreatic cancer. Am J Surg 1996;172(4): 350e2.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
365
[255] Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6(224):224ra224. [256] Januszewicz W, Fitzgerald RC. Early detection and therapeutics. Mol Oncol 2019;13(3):599e613. [257] Cohen JD, Javed AA, Thoburn C, et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci USA 2017;114(38):10202e7. [258] Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science (New York NY) 2018;359(6378): 926e30. [259] Le Calvez-Kelm F, Foll M, Wozniak MB, et al. KRAS mutations in blood circulating cell-free DNA: a pancreatic cancer case-control. Oncotarget 2016;7(48):78827e40. [260] Mellby LD, Nyberg AP, Johansen JS, et al. Serum biomarker signature-based liquid biopsy for diagnosis of early-stage pancreatic cancer. J Clin Oncol 2018;36(28):2887e94. [261] Staal B, Liu Y, Barnett D, et al. The sTRA plasma biomarker: blinded validation of improved accuracy over CA19-9 in pancreatic cancer diagnosis. Clin Cancer Res 2019. clincanres.3310. [262] Colwell J. Illumina spin-off to develop early-detection test. Cancer Discov 2016;6(4):Of3. [263] Zhang W, Xia W, Lv Z, Ni C, Xin Y, Yang L. Liquid biopsy for cancer: circulating tumor cells, circulating free DNA or exosomes? Cell Physiol Biochem 2017;41(2):755e68. [264] Sheng W, Ogunwobi OO, Chen T, et al. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip 2014;14(1):89e98. [265] Nagrath S, Sequist LV, Maheswaran S, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007;450(7173):1235e9. [266] Stott SL, Hsu CH, Tsukrov DI, et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc Natl Acad Sci USA 2010;107(43):18392e7. [267] Wang S, Thomas A, Lee E, Yang S, Cheng X, Liu Y. Highly efficient and selective isolation of rare tumor cells using a microfluidic chip with wavy-herringbone micropatterned surfaces. Analyst 2016;141(7):2228e37. [268] Gleghorn JP, Pratt ED, Denning D, et al. Capture of circulating tumor cells from whole blood of prostate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate-specific antibody. Lab Chip 2010;10(1):27e9. [269] Imperiale TF, Ransohoff DF, Itzkowitz SH, et al. Multitarget stool DNA testing for colorectal-cancer screening. N Engl J Med 2014;370(14):1287e97. [270] Haug U, Wente MN, Seiler CM, Jesenofsky R, Brenner H. Stool testing for the early detection of pancreatic cancer: rationale and current evidence. Exp Rev Mol Diagn 2008;8(6):753e9. [271] Kisiel JB, Yab TC, Taylor WR, et al. Stool DNA testing for the detection of pancreatic cancer: assessment of methylation marker candidates. Cancer 2012;118(10):2623e31. [272] Stool testing for pancreatic cancer. 2019. https://clinicaltrials.gov/ct2/show/ NCT01104129. ClinicalTrials.gov. [273] Previdi MC, Carotenuto P, Zito D, Pandolfo R, Braconi C. Noncoding RNAs as novel biomarkers in pancreatic cancer: what do we know?. 2016. [274] Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA 2005;102(39):13944e9.
366
Theranostic Approach for Pancreatic Cancer
[275] Vicentini C, Fassan M, D’Angelo E, et al. Clinical application of microRNA testing in neuroendocrine tumors of the gastrointestinal tract. Molecules (Basel Switz) 2014;19(2):2458e68. [276] Roldo C, Missiaglia E, Hagan JP, et al. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol 2006;24(29):4677e84. [277] Matthaei H, Wylie D, Lloyd MB, et al. miRNA biomarkers in cyst fluid augment the diagnosis and management of pancreatic cysts. Clin Cancer Res 2012;18(17):4713e24. [278] Li A, Yu J, Kim H, et al. MicroRNA array analysis finds elevated serum miR-1290 accurately distinguishes patients with low-stage pancreatic cancer from healthy and disease controls. Clin Cancer Res 2013;19(13):3600e10. [279] Ruebel K, Leontovich AA, Stilling GA, et al. MicroRNA expression in ileal carcinoid tumors: downregulation of microRNA-133a with tumor progression. Mod Pathol 2010;23(3):367e75. [280] Li SC, Essaghir A, Martijn C, et al. Global microRNA profiling of well-differentiated small intestinal neuroendocrine tumors. Mod Pathol 2013;26(5):685e96. [281] Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov 2006;5(3):219e34. [282] Halbrook CJ, Pontious C, Kovalenko I, et al. Macrophage-released pyrimidines inhibit gemcitabine therapy in pancreatic cancer. Cell Metab 2019;29(6):1390e1399.e6. [283] Terry S, Savagner P, Ortiz-Cuaran S, et al. New insights into the role of EMT in tumor immune escape. Mol Oncol 2017;11(7):824e46. [284] Diehn M, Cho RW, Lobo NA, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009;458(7239):780e3. [285] Kong B, Qia C, Erkan M, Kleeff J, Michalski CW. Overview on how oncogenic Kras promotes pancreatic carcinogenesis by inducing low intracellular ROS levels. Front Physiol 2013;4:246. [286] Vassalli G. Aldehyde dehydrogenases: not just markers, but functional regulators of stem cells. Stem Cells Int 2019;2019:3904645. [287] Dehghan Harati M, Rodemann HP, Toulany M. Nanog signaling mediates radioresistance in ALDH-positive breast cancer cells. Int J Mol Sci 2019;20(5). [288] Croker AK, Allan AL. Inhibition of aldehyde dehydrogenase (ALDH) activity reduces chemotherapy and radiation resistance of stem-like ALDHhiCD44(þ) human breast cancer cells. Breast Cancer Res Treat 2012;133(1):75e87. [289] Cojoc M, Mabert K, Muders MH, Dubrovska A. A role for cancer stem cells in therapy resistance: cellular and molecular mechanisms. Semin Cancer Biol 2015;31:16e27. [290] Tsubouchi K, Minami K, Hayashi N, et al. The CD44 standard isoform contributes to radioresistance of pancreatic cancer cells. J Radiat Res 2017;58(6):816e26. [291] Hurt EM, Kawasaki BT, Klarmann GJ, Thomas SB, Farrar WL. CD44þ CD24() prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis. Br J Cancer 2008;98(4):756e65. [292] Wang S, Huang S, Sun YL. Epithelial-mesenchymal transition in pancreatic cancer: a review. BioMed Res Int 2017;2017:2646148. [293] A phase 1/2 safety study of intratumorally dosed INT230-6. 2019. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03058289?term¼NCT03058289&rank¼1.
Recent advances in molecular diagnostics and therapeutic targets for pancreatic cancer
367
[294] Intensity therapeutics highlights INT230-6 data in advanced solid tumors at society for immunotherapy of cancer’s (SITC) 33rd annual meeting. Intensity Therapeutics; November 8, 2018. https://intensitytherapeutics.com/intensity-therapeuticshighlights-int230-6-data-in-advanced-solid-tumors-at-society-for-immunotherapyof-cancers-sitc-33rd-annual-meeting/. [295] El-Khoeuiry A, Siu LL, Azad N, et al. 1160P Phase I/II evaluation of intratumoral INT230-6 for the treatment of solid tumors. Ann Oncol 2018;29(Suppl. 8). [296] Sun Q, Ojha T, Kiessling F, Lammers T, Shi Y. Enhancing tumor penetration of nanomedicines. Biomacromolecules 2017;18(5):1449e59. [297] Ye J, Liu E, Yu Z, et al. CPP-assisted intracellular drug delivery, what is next? Int J Mol Sci 2016;17(11).