Stories of drug repurposing for pancreatic cancer treatment—Past, present, and future


Acknowledgments

We want to apologize to all authors whose work could not be cited due to space restrictions/limitations in this chapter.

Introduction

Cancer in general is one of the leading causes of mortality worldwide [ ]. Pancreatic ductal adenocarcinoma (PDAC), as the most frequent malignant tumor of the pancreas, has not a high incidence on a global level. However, it is projected to become the second leading cause of cancer-related death in particular in the United States and in Europe by 2030 [ ]. At the time of diagnosis, only about 10% of all PDACs are local disease with a 32% 5-year survival rate and 29% are regional disease with a 12% 5-year survival rate [ ] Some of the latter group will become eligible for surgery after upfront chemotherapy. Unfortunately, the majority of patients (52%) are diagnosed with distant stage disease and will undergo relatively inefficient systemic therapy regimens, such as modified FOLFIRINOX (FFX; fluorouracil, folinic acid, irinotecan, oxaliplatin) or gemcitabine (GEM) plus nanoparticle albumin-bound paclitaxel (nab-paclitaxel; GnP) with a 5-year survival rate of currently only 3% at the moment [ , ]. Although lately, some progress can be seen, due to the mentioned facts, PDAC patients usually still encounter a dismal prognosis at the time of diagnosis, and thus, the 5-year overall survival (OS) of all PDAC patients is about 8% [ ]. Prevention is difficult in this neoplasm, where only few factors associated with higher incidence are known and only a small number of specific risk factors are tightly linked to its occurrence. Randomized screening of patients at high risk, such as those with a family history of PDAC, by MRI or endoscopic ultrasound has not yielded the expected success [ ]. Similarly, current screening methods do not permit early discovery, which is in part due to the difficult accessibility of the pancreas, although tremendous efforts are invested into approaches that might help to delineate the disease at earlier, and therefore, treatable stages of disease, such as analysis of exosomes or other liquid biomarkers [ ].

At the same time, remarkable energy is being invested in drug discovery, not only for PDAC but also anticancer strategies in general. Research spending of private and public foundations (e.g., National Institutes of Health, Deutsche Forschungsgemeins, etc.) is rising, biotechnology companies or spin-offs are being launched, and established pharmaceutical companies are investing increasingly higher budgets on drug development. Many of those ventures have not translated into more or better drugs to the extent of the invested resources. Estimates of different studies show that research and development expenditures for a new drug might accumulate up to $2.6 billion or higher [ ]. Moreover, from basic research to the eventual FDA filing, it is a lengthy process and might take anywhere from 9 to 20 years ( Fig. 9.1 ). Apart from the economic and long-lasting efforts, respectively, high failure rates add up to a very interminable route, and so, overall, only about 5%–12% of all drugs that enter Phase I clinical trials ultimately receive FDA approval ( Figs. 9.1 and 9.2 ) [ , ].

Figure 9.1, Illustration of the estimated costs and time that is currently estimated to be needed for the development of a drug from bench (basic research) to bedside (FDA approval). Repurposing significantly shortens that trajectory (upper image). Transition probability for clinical trials based on different phases as well as overall transition (modified according to DiMasi and colleagues, 2016 [ 10 ]).

Figure 9.2, Challenge models of mouse avatars, patient-derived xenografts (PDX), or patient-derived organoids (PDO) with mutation profiling of patients as well as drugs. Together, this could transform into a bioinformatics pipeline with gold standard methods [ 20 ].

Drug repurposing is therefore an increasingly attractive field for alternative drug development. This is especially appealing because, in general, clinical trials are designed to test very specific questions and narrow hypotheses but cannot focus on and/or expose unexpected benefits [ ]. Moreover, patients who might profit in a different way might not even be included in initial trials, e.g., children, pregnant women, or elderly people. The latter two issues could explain the high failure rate in clinical trials in part. On the other hand, estimates attribute secondary indications to about 90% of all approved drugs because of molecular origins that different diseases have in common, which makes it highly attractive to “teach new tricks to old dogs” [ ]. In this regard, drug repurposing or its alternative terms (“new uses for old drugs,” “drug repositioning,” “drug reprofiling,” “therapeutic switching”) have gained considerable attention not only in the field of cancer but also in medicine in general. In summary, this terminology in principal refers to identifying or developing new uses for existing or abandoned pharmacotherapies [ ]. In this chapter, we will briefly summarize common strategies toward drug repurposing in general and discuss successful examples in medicine. In the main part, we focus on repurposing approaches in the past, present, and future in PDAC. For the scope of this chapter, medical device repurposing will not be discussed.

General strategies toward drug repurposing

To discover new purposes for established drugs, different routes have been taken historically. The first approach relies on ongoing basic research and scientific discovery of novel mechanisms of action. In this regard, after the introduction of a drug or device, investigations might reveal the dependence of a different tumor on the same pathway that could then lead to the off-label use of a previously well-established drug. A prime example for that is the expansion of imatinib, originally developed for the treatment of chronic myelogenous leukemia (CML) to other cancer types on the basis of common principal pathways [ , ].

Second, a translational way of discovery depends on the understanding of the pathophysiologic basis of how a drug works. With that, scientists can widen the search for additional applications of a drug to completely different conditions with yet the same mechanism. For instance, beta-adrenergic antagonists or beta-blockers, originally developed to treat to treat angina and cardiac arrhythmias [ , ], have also been applied to alleviate performance anxiety or essential tremor [ ].

Third, clinical observations might help to understand additional and unrecognized (positive) side effects of FDA-approved drugs. For example, bupropion use could be linked to easing smoking cessation but was originally developed as an antidepressant drug [ ]. In a similar way, sildenafil was developed to treat angina pectoris and arterial hypertension. During early phase studies, it was discovered that erectile dysfunction could be significantly improved by its application [ ].

Combining the abovementioned pathways with cutting-edge techniques, such as high-throughput screening as well as computational approaches, might lead to reasonable and modern ways to repurpose drugs to other diseases or vice versa. Ideally, Connectivity Maps of drugs or even drug pipelines, diseases and their most abundant pathways, and genes of the respective tumor would be created. The obtained data could be challenged against each other, evaluated with phenotypic screening in modern models of disease (among others, patient-derived xenografts [PDX], mouse avatars or patient-derived organoids [PDO]), and personalized candidate drugs would be proposed by bioinformatics and/or artificial intelligence (AI) ( Fig. 9.2 ). In this regard, PDOs have the advantage of accelerated assessment of individual neoplastic cell dependencies and drug responses of the pipeline. Also, they can be transplanted into mice with various genetic backgrounds to investigate interactions with the TEM [ ]. Genetically engineered mouse models, including CRISPR/Cas9-edited animals, could serve as avatars to novelties of single and combination therapies as suggested by the drug pipeline [ ]. Genomic alterations often prioritize driver pathways on which the tumor becomes dependent. This dependence can—in theory—be exploited for significant cancer viability inhibition. Moreover, genomic data or gene expression signatures could help to refine data and hence classify additive effects of potential combinations of clinically useful treatment options by prioritizing therapeutic vulnerabilities or helping to identify suitable biomarkers.

Perspectives of drug repurposing and successful examples

Recent history of drug repurposing is rich in examples of successfully repurposed drugs including some with initial failure or even severely harmful side effects.

In the field of cardiology, several blockbuster drugs have been developed to help people with different ailments such as clogged coronary arteries, arrhythmia, or arterial hypertension. In this regard, the calcium channel blocker verapamil was introduced in 1963 for the treatment of angina pectoris; however, through basic and translational strategies as described in Section General strategies toward drug repurposing , pathologies in calcium channel functioning were shown to be involved in many other medical conditions, such as esophageal spasms, peripheral vascular disease, preterm labor, hypertension, and even migraine. For all of those ailments, verapamil is in use nowadays in a repurposed manner [ ]. Another prime example in the field of cardiology is the widened use of beta-blockers, initially introduced to treat tachycardic conditions, but with remarkable effects for patients with performance anxiety or essential tremor [ ]. Not only that, but as we will discuss later, beta-blockers might also be very useful in the field of oncology to treat tumors that thrive on sympathetic, adrenergic signaling networks [ ].

Last, the phosphodiesterase inhibitor sildenafil (Viagra) was originally developed for coronary artery disease and angina pectoris, which was ineffective. However, clinical testing showed relevant side effects that were fully exploited by repurposing of the drug in order to improve male erectile disfunction and pulmonary arterial hypertension. It has become a blockbuster drug of its own ever since its release in 1998 [ ].

A notable example of initial failure having at first had teratogenic side effects to unborn babies is thalidomide. Initially developed as a sedative drug in the early 1960s, it proved to cause severe malformations if taken during pregnancy [ ]. Nevertheless, basic, translational, and clinical scientific approaches showed its clinical benefits in treating leprosy (FDA approval in 1998) and mechanistically through inhibition of TNF-α as an antitumorigenic drug for multiple myeloma as well as rheumatoid arthritis [ ].

Many other interesting examples of successful drug repurposing are summarized in Table 9.1 below.

Table 9.1
Successfully repurposed blockbuster drugs and their original purpose.
Drug Original purpose Repurpose References
Beta-blockers Tachycardia Performance anxiety or essential tremor
Bupropion (Wellbutrin) Depression Smoking cessation [ ]
Diclofenac Rheumatoid arthritis, osteoarthritis Analgesia, primary dysmenorrhea, ankylosing spondylitis
Duloxetine (Cymbalta) Depression Stress urinary incontinence
Everolimus mTOR,
Renal, astrocytoma immunosuppression
Imatinib (Gleevec, Glivec) CD117 = cKIT
Chronic myelogenous leukemia (CML)
Gastrointestinal stroma tumors (GIST) [ , ]
Sildenafil Coronary artery disease Erectile dysfunction
Pulmonary arterial hypertension
[ ]
Thalidomide Sedative drug in the early 1960s → teratogenic effects Leprosy, 1998 inhibiton of TNF-α → tumors (multiple 2006 myeloma), rheumatoid arthritis [ ]
[ ]https://www.fda.gov/AboutFDA/CentersOffices/CDER/ucm095651.htm.
Verapamil Ca 2+ channel blocker verapamil (introduced in 1963) for angina pectoris Esophageal spasms, peripheral vascular disease, preterm labor, hypertension, prophylaxis to migraine [ ]

Drug repurposing strategies in pancreatic cancer (PDAC)

This section reviews strategies that are being applied to the specific purpose of identifying helpful drugs for developing novel approaches toward PDAC treatment. We include trends of both academic institutions and pharmaceutical industry and try to corroborate our statements with prominent case studies of past and present.

Interestingly, exploration of scientific search engines revealed only a small number of hits using the search terms “drug repurposing” and “pancreatic cancer” or “drug repositioning” and “pancreatic cancer” ( Fig. 9.3 ), which emphasizes the enormous potential that these approaches might still promise. However, even in the field of pancreatic cancer, an increase in publications over the last 3–4 years could be noticed and we have reviewed some of those trends before ( Fig. 9.3 ) [ , ].

Figure 9.3, Illustration of the number of publications per year. A steady increase was detected with most of the publications published after 2016. In purple numbers found in Web of Science and in gray numbers found in PubMed databases are illustrated. Terms for the search were as follows: “drug repurposing” and “pancreatic cancer.”

Last, this section will include a subjective outlook on future repurposing strategies and fields that we consider promising with substantial opportunities of improving medical treatment for patients with PDAC.

Case studies of the past

Permanent intense scientific efforts are undertaken and try to seek more curative treatment options for PDAC in adjuvant, neoadjuvant, or primary metastatic settings. However, most efforts remain unsatisfactory, making PDAC one of the leading causes for cancer-related death in Europe and the United States [ , ]. Still, resection of the primary tumor is the only curative option that is possible in roughly 20% of all PDAC patients, and surgery increases the 5-year survival rate significantly [ , ]. Conversely, locally unresectable tumors, concurrent distant metastasis, or local recurrence render patient prognosis devastating [ ].

The search for further and more potent therapeutic approaches is imperative and should therefore include both the search for novel drugs as well as repurposing compounds that have already shown antitumoral activity in other cancers [ ].

In this section, we aim to describe case studies of drugs repurposed in the past, their original mechanism of action, and their implication in PDAC therapy and reveal how they performed.

CSC pathway inhibition

The concept of cancer stem cells (CSCs) postulates that in heterogeneous tumors, a subgroup of cells that are not necessarily predefined but endowed with CSC characteristics, could sit at the top of hierarchy. Initially portrayed as a subclass of indefinitely propagating neoplastic cells that produce an overt cancer (self-renewal and tumorigenesis) [ ], current literature ascribes many other properties including a higher grade of plasticity to CSCs. This means on the one hand that different CSC subcategories could exist and on the other hand that non-CSCs can potentially dedifferentiate into CSCs presumably by activation of cancer stemness-associated signaling pathways such as WNT/RSPO [ , ], Nodal/Activin [ ], miR-based pathways [ ], or Notch. Most of them can control epithelial–mesenchymal transition (EMT) in PDAC and are therefore very potent mechanisms that regulate steps of invasive and metastatic cascades [ ]. These signaling pathways, moreover, are regulated by feedback loops in CSCs, e.g., the miR-17-92 cluster can affect p38alpha and subsequently canonical WNT signaling in lung cancer [ ], which in turn regulates or is regulated by ERK [ ]. These interplays suggest a highly orchestrated and controlled interaction in supposedly deregulated cancer cells that offer unique opportunities to specifically target those CSCs with previously established FDA-approved drugs. Cycling between the different states of stemness and differentiation means that cells must be able to change their functional properties. Apart from the mentioned signaling pathways, metabolic dependencies seem to profoundly impact the plasticity of CSCs'. Preclinical models of CSCs in PDAC were able to demonstrate that FDA-approved drugs could possess inhibitory properties to the abovementioned CSC-specific capabilities. Here, we will summarize mostly preclinical data around repurposing FDA-approved drugs that could confront PDAC and its CSCs. Although the concept of CSCs is not completely novel anymore, given the ongoing challenges of a rising PDAC incidence, repurposing drugs might emerge as a considerable opportunity with new perspectives and angles in the course of PDAC treatment.

Canonical WNT signaling

Canonical WNT signaling is activated by the binding of glycoproteins (WNTs) to their coreceptors LGR5/6 and frizzled (FZD). This signal is then transduced into the cell where beta-catenin serves as its transcription factor [ ]. It is also known that the RSPO/LGR complex can robustly enhances a preexisting WNT signal. LGR5, in particular, acts as a marker for normal stem cells [ ] and CSCs [ ]. Thus, the WNT/RSPO/LGR pathway comprises a very intricate system with multiple members and critical importance in CSCs, predominantly in colorectal [ , ] and liver cancer [ ]. Liver and colorectal cancers are inherently hooked to canonical WNT signaling due to their activating mutations in the pathway (APC and beta-catenin genes). Inhibition of the pathway on the other hand is everything but trivial because of its complex and intertwined structure; various studies report successful attempts, e.g., with salinomycin-downregulated WNT signaling in CSCs of chronic lymphatic leukemia (CLL) [ ]. Another very intriguing agent is the FDA-approved small molecule inhibitor aprepitant, which is directed against the NK1-receptor (NK1-R). Its primary clinical use is for chemotherapy-induced nausea and vomiting (CINV), but it also showed strong and efficient inhibitory activity in hepatoblastoma in vitro and in vivo [ ]. Aprepitant as well as similar NK1-R inhibitors robustly decreased the viability of CSCs in colon and pediatric liver cancer (hepatoblastoma) [ ] by potently suppressing both AKT/mTOR and canonical WNT [ ] through the disruption of the FoxM1/beta-catenin complex [ ]. Intriguingly, FoxM1 is often deregulated in PDAC and exerts metabolic control over tumor growth influencing oxidative glycolysis (Warburg effect) [ ]. Last, low-dose ketamine induced ADNP expression, which in turn acted as a WNT repressor in a colorectal cancer model. This ultimately inhibited tumor growth and prolonged survival of tumor-bearing mice in vivo [ ].

Disulfiram

Disulfiram is a compound that is used to treat patients with chronic alcoholism. However, it is also known to modify a range of cellular functions, some of which could be useful to target in PDAC. Notably, disulfiram seems to have an effect on pancreatic CSCs by elimination of the highly tumorigenic and therapy-resistant Aldefluor-positive cells in vitro [ ]. Additionally, EMT was significantly inhibited by inhibition of ERK and NF-
κ
B signaling in breast cancer cells [ ].

In a Phase 2 clinical pilot study ( NCT03714555 ) involving metastatic PDAC, disulfiram and copper gluconate will be added to chemotherapy (nab-paclitaxel plus GEM, FOLFIRINOX, or GEM mono) [ ]. Additionally, disulfiram is a potential inhibitor of muscle degeneration and as such seems to ameliorate muscle wasting in animal models. In this regard, it could exert supplementary effects in patients with cancer cachexia or sarcopenia, which are conditions that not only debilitate the PDAC patients early on in the course of the disease but also result in chemotherapy-precluding frailty. Moreover, the use of disulfiram was supposed to sensitize cancer cells to GEM hydrochloride ( NCT02671890 ) [ ].

Quinomycin A

The quinoxaline antibiotic quinomycin A was attributed with a potential to influence several important CSC hallmarks, among them, inhibition of spheroid formation of PDAC, downregulation of several CSC markers, and suppression of the expression of multiple members of the Notch pathway in vitro. Quinomycin A treatment significantly reduced tumor burden in vivo, which suggested that it could be useful as a potent drug to target CSCs in PDAC [ ].

Modulation of epithelial–mesenchymal transition

EMT is highly conserved and was shown to be mechanistically involved in regulating CSC plasticity of different tumor types, e.g., breast cancer, colorectal cancer, and PDAC [ , ]. Given that background, it was hypothesized that inhibition of EMT should then diminish the CSC population of a tumor and, in consequence, decrease the frequency of tumor relapse and metastasis. In light of this, the development of strategies to inhibit EMT has been pursued. In a systematic drug screen for EMT inhibitors, Meidhof and colleagues were able to show that mocetinostat, a class I histone deacetylase inhibitor, can act as an epigenetic drug by interfering with ZEB 1 function. This restored miR-203 expression and ultimately repressed EMT as well as CSC properties in PDAC and prostate cancer. In addition, mocetinostat was also able to resensitize CSCs for treatment with conventional chemotherapy [ ]. In PDAC, therapy resistance, e.g., against GEM as an established chemotherapeutic agent for systemic treatments, seems to depend heavily on EMT mechanisms. It was shown that inhibition of EMT increased the expression of two types of human nucleoside transporters, namely the concentrative nucleoside transporter and the equilibrative nucleoside transporter (ENT) proteins, which are important for the molecular transport of hydrophilic GEM into the cells [ ]. However, invasive or metastatic behaviors of PDAC were not significantly changed by the same treatment. Together, that might suggest that targeting EMT in PDAC could be of therapeutic benefit affecting various CSC hallmarks downstream of EMT [ ].

Plerixafor

CXCR4, a receptor to CXCL12 (SDF1) or MIF, was initially found to be mainly expressed on hematopoietic stem cells, and plerixafor (Mozobil) was developed to inhibit CXCR4. In this regard, plerixafor is in use to mobilize stem cells, e.g., before harvest for transplantation therapies. Besides from that, CXCR4 was also found to be highly expressed in cells with increased invasive and metastatic capabilities in PDAC and other malignancies [ , ]. Moreover, CXCR4 conferred drug resistance in some cell populations [ ]. With that, plerixafor could be useful in targeting cancer, especially in the context of highly CXCL12-producing surroundings. Last, current studies showed that it might actually be supportive in combination with immunotherapeutic drugs such as α-PD-1/α-CTLA4 [ ].

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here