Purpose:

Iododeoxyuridine (IUdR) is a potent radiosensitizer; however, its clinical utility is limited by dose-limiting systemic toxicities and the need for prolonged continuous infusion. 5-Iodo-2-pyrimidinone-2′-deoxyribose (IPdR) is an oral prodrug of IUdR that, compared with IUdR, is easier to administer and less toxic, with a more favorable therapeutic index in preclinical studies. Here, we report the clinical and pharmacologic results of a first-in-human phase I dose escalation study of IPdR + concurrent radiation therapy (RT) in patients with advanced metastatic gastrointestinal (GI) cancers.

Patients and Methods:

Adult patients with metastatic GI cancers referred for palliative RT to the chest, abdomen, or pelvis were eligible for study. Patients received IPdR orally once every day × 28 days beginning 7 days before the initiation of RT (37.5 Gy in 2.5 Gy × 15 fractions). A 2-part dose escalation scheme was used, pharmacokinetic studies were performed at multiple time points, and all patients were assessed for toxicity and response to Day 56.

Results:

Nineteen patients were entered on study. Dose-limiting toxicity was encountered at 1,800 mg every day, and the recommended phase II dose is 1,200 mg every day. Pharmacokinetic analyses demonstrated achievable and sustainable levels of plasma IUdR ≥1 μmol/L (levels previously shown to mediate radiosensitization). Two complete, 3 partial, and 9 stable responses were achieved in target lesions.

Conclusions:

Administration of IPdR orally every day × 28 days with RT is feasible and tolerable at doses that produce plasma IUdR levels ≥1 μmol/L. These results support the investigation of IPdR + RT in phase II studies.

Translational Relevance

5-Iodo-2-pyrimidinone-2′-deoxyribose (IPdR) is a prodrug of iododeoxyuridine (IUdR), a halogenated thymidine analog that is a well-recognized radiation-sensitizing drug, with the degree of radiosensitization correlated directly with plasma levels and IUdR-tumor cell DNA incorporation. The clinical use of IUdR is limited by administration issues and moreover, by dose-limiting bone marrow and gastrointestinal toxicities. This report of the first-in-human tolerance study of IPdR confirms that IPdR is predictably and readily absorbed following oral administration, is metabolized to IUdR principally by hepatic aldehyde oxidase, and demonstrates pharmacokinetics that compare favorably to those of IUdR. In this dose-finding pharmacokinetic phase I trial, IPdR was administered orally, once every day for 28 days, with concurrent radiation therapy (RT) to patients receiving palliative RT for advanced gastrointestinal cancers. IPdR was found to be tolerable at doses that produced plasma IUdR levels that have previously been associated with clinically significant radiosensitization, supporting further testing of IPdR as a potential radiosensitizing agent.

Radiation therapy (RT) is a critical component of cancer therapy, in both the curative and palliative settings, with approximately 60% of patients receiving RT at some time during the course of their cancer treatment (1). The availability of additional radiosensitizing drugs that improve the efficacy/toxicity profile of RT would represent a meaningful advance in cancer therapy, potentially improving survival and lessening morbidity for patients with cancer in a wide range of clinical situations. Indeed, the development of radiosensitizers has been targeted as a priority in a collaborative project involving the NCI, the Radiation Therapy Oncology Group (RTOG; now part of NRG Oncology Group) and investigators from the United Kingdom (2, 3).

A number of systemic agents are used in clinical practice to enhance the effect of RT (2), including platinum compounds (4–6), signal transduction inhibitors (7), fluoropyrimidines (8–11), gemcitabine (12), and temozolomide (13). The use of cytotoxic agents with RT has been found to benefit patients with select tumor types, for example temozolomide in glioma, fluoropyrimidines in gastrointestinal (GI) malignancies, and cetuximab/EGFR for head and neck cancers. Although attempts have been made to exploit tumor hypoxia to improve radiation response, no agents in this class are currently considered standard-of-care (14). Broader use of these agents has been limited by toxicity, especially in the context of multimodality, multiagent treatment regimens. These existing examples validate the clinical proof-of-principle of radiosensitization and encourage development of additional radiosensitizing agents with higher therapeutic indexes.

5-Iodo-2-pyrimidinone-2′-deoxyribose (IPdR) is a prodrug of iododeoxyuridine (IUdR), a pyrimidine analog that has been recognized as a radiosensitizing agent since the early 1960s (15, 16). IPdR offers several key advantages compared with IUdR, including a more favorable therapeutic index.

The mechanism of IUdR-mediated radiosensitization involves the cellular uptake and metabolism of IUdR through the thymidine salvage pathway. IUdR undergoes initial intracellular phosphorylation to the monophosphate derivative by the rate-limiting enzyme, thymidine kinase, followed by sequential phosphorylation to the triphosphate. This modified analog is then utilized (i.e., incorporated) by DNA polymerase during scheduled (S-phase) and unscheduled DNA synthesis, in competition with thymidine triphosphate. DNA incorporation is needed for radiosensitization by ionizing radiation (IR), in human tumors as well as normal cells, and the extent of radiosensitization correlates directly with the %IUdR DNA replacement (17–19). IR-mediated radiosensitization by IUdR results in the generation of highly reactive uracil free radicals by IR, which may also damage unsubstituted complementary-strand DNA, resulting in increased DNA single strand breaks and increased double-strand breaks (18–20). It is also known that the repair of IR damage may also be reduced by pre-IR exposure to IUdR. IUdR is rapidly metabolized in both rodents and humans when given as a bolus infusion with a plasma half-life of <5 minutes (15). Consequently, prolonged continuous drug infusion over several weeks before and during RT is necessary to maximize the proportion of tumor cells that incorporate IUdR during the S phase of the cell cycle (21, 22).

In the 1980 to 1990s, clinical trials of IUdR as a radiosensitizer were performed, focusing on patients presenting with high-grade gliomas (23–26) and sarcomas (27, 28). These studies demonstrated that the magnitude of radiosensitization correlates directly with plasma IUdR levels and the %IUdR-DNA tumor cellular replacement, and that both measurements can serve as radiosensitization biomarkers (29, 30). Additionally, in small series of patients with head and neck cancers or liver metastases from colorectal cancer, the %IUdR-DNA cellular incorporation in tumors ranged to 5%, but was less than 1% in adjacent normal liver tissue, further supporting a therapeutic window for IUdR-mediated radiosensitization (30–32). Although IUdR has clear activity as a radiosensitizer, it requires prolonged continuous infusion before and throughout the course of RT, and systemic toxicities (myelosuppression, GI toxicities) limit the tolerable dose and thus IUdR's potential for radiosensitization. These issues have constrained the development of IUdR as a radiosensitizer (23–26).

IPdR is a prodrug of IUdR, which is administered orally and then efficiently converted to IUdR by a hepatic aldehyde oxidase (33). Although preclinical studies have many limitations, the broad range of pharmacology and safety testing in mice, rats, ferrets, and Rhesus monkeys has been promising in terms of the ability of IPdR to mediate radiosensitization with acceptable toxicity (34–38). Strong support for advancement of IPdR to the clinic was provided by radiosensitization studies in vivo using human tumor xenograft models: 2 colorectal cancer models (HT29, HCT-116), and 2 glioblastoma (GBM) cell lines (U251 and U87; refs. 34, 35, 37, 39, 40).

This collection of preclinical studies demonstrated that: (i) IPdR could be administered and tolerated orally (gavage) using multiple daily dosing up to 28 days; (ii) IPdR is efficiently and predictably metabolized into the active metabolite, IUdR; (iii) relatively prolonged plasma levels of IUdR can be achieved; (iv) %IUdR-DNA incorporation was demonstrated to be 2 to 3 times greater in proliferating tumor and 2- to 3-fold less in proliferating normal tissues (bone marrow and GI epithelium) compared with continuous infusion IUdR at the MTD; (v) radiation sensitizer enhancement ratios of 1.3 to 6 were achieved using human cancer xenograft models, and compared favorably with those of continuous infusion IUdR at the MTD (≤1.1); and (vi) systemic toxicity was manageable at effective doses. These data were used to support an Investigational New Drug Application (IND; #70,333) and to conduct an initial phase 0 trial of single-dose IPdR in patients with advanced malignancies that demonstrated radiosensitizing plasma levels of IUdR could be achieved following oral administration of IPdR (41).

The choice of patient population for this phase I and pharmacology trial was based on an NCI-sponsored IPdR Experts Conference that prioritized GI cancers and the NCI Colorectal Cancer Working Group's interest in combining IPdR with standard-of-care therapy for patients with rectal cancer (11). Patients undergoing intermediate-dose palliative RT for metastatic GI cancers received a single every day oral dose of IPdR beginning 7 days before the initiation of RT through the final day of RT. Plasma levels of IPdR, IUdR, and metabolites were measured at multiple time points throughout the 28 days, patients were monitored for toxicity, and tumor response was assessed at Week 8. The maximum tolerated dose (MTD) and the recommended phase II dose (RP2D) were established in this patient population. Results of this trial support phase II testing of oral IPdR as a radiosensitizing agent.

Patient population

Patients 18 years and older with histologically or cytologically confirmed advanced, incurable cancers of the esophagus, liver, stomach, small bowel, pancreas, bile duct, colon, or rectum referred for palliative RT to the chest, abdomen, and/or pelvis were eligible for the trial. Patients received no other systemic therapy for 1 month prior to study and for the duration of study (8 weeks). Patients receiving palliative RT for advanced primary tumors or metastatic disease were eligible. Eligible patients had acceptable organ function based on laboratory data and an Eastern Cooperative Oncology Group performance status (ECOG PS) of ≤2. Patients must not have received prior RT to the tumor site being irradiated on this study. Exclusion criteria included brain metastases, a history of allergic reactions to compounds of similar chemical or biologic composition to IPdR, uncontrolled intercurrent illness, psychiatric illness/social situation that would limit compliance with study requirements and pregnancy or breast feeding. The trial was conducted under a NCI-sponsored IND (#70,333) and conducted by the Brown University Oncology Group (BrUOG) at Lifespan Rhode Island Hospital (RIH). The protocol and informed consent documents were approved by the Cancer Therapy Evaluation Program (CTEP) and the Lifespan-RIH IRB. Patients provided written informed consent prior to enrollment. Protocol design and conduct followed all applicable regulations, guidances, and local policies. The study is registered on ClinicalTrials.gov, NCT02381561.

Trial design and treatment

This was a single-institution, single-arm dose escalation study. IPdR was supplied by the Developmental Therapeutics Program (DTP), NCI. Patients received a single every day oral dose of IPdR, beginning 7 days prior to the initiation of RT and continuing QD through the final day of RT (Days 1–7 without RT; Days 8–28 with RT; Fig. 1). IPdR was supplied as 75 and 300 mg capsules. Capsules were taken on an empty stomach (i.e., NPO for 2 hours prior to and 1 hour following oral administration). On the days of RT, IPdR was administered within 0.5 to 2 hours prior to RT. Patients kept a medication log, recording the date, time and number of capsules of each dose.

Figure 1.

Schema. Phase I and pharmacology trial of oral IPdR as a prodrug for IUdR-mediated radiosensitization in patients receiving palliative RT for metastatic GI cancers (NCT02381561).

Figure 1.

Schema. Phase I and pharmacology trial of oral IPdR as a prodrug for IUdR-mediated radiosensitization in patients receiving palliative RT for metastatic GI cancers (NCT02381561).

Close modal

Patients received intensity modulated radiation therapy (IMRT; 4–18 MeV photons) to a total dose of 37.5 Gy using 15 2.5 Gy fractions delivered once daily on a Monday to Friday schedule during Weeks 2 to 4 of IPdR treatment. This intermediate-dose RT schedule is not typical for the palliative setting, where more commonly, a total dose of 30 Gy is delivered in 10 3 Gy fractions or 20 to 25 Gy is administered in 4 to 5 Gy fractions, and where 3-D planning is often the norm. The choice of RT dose, schedule and modality used in this trial reflected the goals of appropriate patient care and the generation of results that could be applied to the clinical setting of the use of RT for GI cancers. IMRT was used to limit the dose to adjacent normal tissues and to best quantitate any local (within the 50% isodose volume) toxicities of IMRT + IPdR using total radiation doses equivalent to those used in the pre- and/or postoperative settings for GI cancers (45–50.4 Gy over 5 to 6 weeks). The IMRT treatment plan for each patient was based on an analysis of the volumetric dose, including dose volume histogram (DVH) analyses of the planning tumor volume (PTV), defined as the clinical tumor volume (CTV) with a 7 mm margin. Ninety percent of the PTV received 95% of the prescribed dose, and 99% of the CTV received 95% of the prescribed dose. The gross tumor volume (GTV) was defined as the symptomatic metastatic or locally recurrent tumor based on contrast enhanced CT or MRI scans, and was expanded by 1.5 cm to define the CTV. The Rhode Island Hospital Department of Radiation Oncology is credentialed for IMRT use in the chest, abdomen, and pelvis through Imaging and Radiation Oncology Core (IROC) Houston for all Eastern Cooperative Oncology Group-American College of Radiology Imaging Network (ECOG-ACRIN), Radiation Therapy Oncology Group (RTOG), and Gynecologic Oncology Group (GOG) protocols (now NRG Oncology).

A primary objective of the study was to determine the safety, toxicities, and MTD of IPdR administered with concurrent RT. Clinical toxicities were graded according to revised NCI Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (version 5.0 when it became available). The study was designed with a 2-part dose escalation scheme. The starting dose was 150 mg every day x 28 days (≅1/10th MTD in ferrets; ref. 38). Part I was a single patient dose escalation of PO IPdR every day x 28 days, with 100% dose escalation for each sequential patient until a treatment related grade 2 systemic toxicity was observed. Three additional patients were then entered at this dose level. If a second patient of these 3 patients experienced a treatment-related grade 2 systemic toxicity (or if a single patient developed treatment related ≥grade 3 toxicity), then a stop/switch rule was invoked for part I, and part II of the dose escalation scheme commenced. If not, the dose was incremented for the next patient. In part II, 3 patients were treated at the final part I dose level, with a 40% dose increment schedule for subsequent dose levels. If 0 of 3 patients did not experience a ≥grade 3 treatment-related dose-limiting toxicity (DLT), accrual continued with 3 patients per dose level, until a ≥grade 3 treatment-related DLT was observed. DLT was reached when any 2 grade 3, systemic or local (within the 50% isodose volume of the radiation plan), treatment-related toxicities were observed in 2 of the up to 6 patients enrolled at that dose level. One additional patient was treated at the dose level below the dose associated with DLT to establish the MTD. The MTD was defined as the dose below which 2 or more of up to 6 patients experience DLT.

Pharmacokinetic studies

For the pharmacokinetic studies, 5 mL of blood was collected at 0, 30, 60, 120, and 240 minutes following the oral dose of IPdR on Days 1, 15, and 22, and a Day 1 24-hour sample was obtained immediately prior to the Day 2 dose. Following collection of blood, plasma was separated via centrifugation, and then shipped per protocol to the NCI Division of Cancer Treatment and Diagnosis (DCTD) Pharmacokinetics Laboratory at the Frederick National Laboratory for Cancer Research in Maryland. Procedures for pharmacokinetic analyses are available in Supplementary Materials and Methods.

In addition to internal review, external review of the raw data was conducted by Theradex and RTI Health Solutions in the context of generating the formal Clinical Study Report, which was reviewed by NCI-CTEP physicians.

Patients

Nineteen patients were enrolled on the trial. Patient demographics and characteristics are presented in Table 1.

Table 1.

Patient demographics and characteristics

Patient characteristic
Age (years) 
 Median 64 
 Range 37–94 
Sex 
 M 11 
 F 
Race 
 Asian 
 Black or African American 
 White 17 
Ethnicity 
 Hispanic or Latino 
 Not Hispanic or Latino 18 
ECOG performance status 
 0 
 1 
 2 
Number of prior treatment regimens 
 1–2 
 3–4 10 
 ≥ 5 
Histology of primary tumor 
 Adenocarcinoma 15 
 Cholangiocarcinoma 
 Hepatocellular carcinoma 
 Squamous cell carcinoma 
Site of primary tumor 
 Ampulla 
 Bile duct 
 Colon 
 Esophagus 
 Liver 
 Pancreas 
 Rectum 
Patient characteristic
Age (years) 
 Median 64 
 Range 37–94 
Sex 
 M 11 
 F 
Race 
 Asian 
 Black or African American 
 White 17 
Ethnicity 
 Hispanic or Latino 
 Not Hispanic or Latino 18 
ECOG performance status 
 0 
 1 
 2 
Number of prior treatment regimens 
 1–2 
 3–4 10 
 ≥ 5 
Histology of primary tumor 
 Adenocarcinoma 15 
 Cholangiocarcinoma 
 Hepatocellular carcinoma 
 Squamous cell carcinoma 
Site of primary tumor 
 Ampulla 
 Bile duct 
 Colon 
 Esophagus 
 Liver 
 Pancreas 
 Rectum 

Determination of MTD

Table 2 describes each patient's treatment, toxicity, and response.

Table 2.

Study treatment, toxicity, and response

Primary tumor histologyPrimary tumor siteIPdR dose (mg every day)Target site (RT field)Toxicity (≥grade 2 and attribution ≥ 3a)GradeAttribution to IPdRaTarget tumor responsebNontarget tumor responseb
AC Colon 150 Liver Met    SD SD 
AC Colon 300 Liver Met    SD SD 
AC Pancreas 600c Pancreas tail ALT PD at D 23c PD at D 23c 
    AST   
    rash   
AC Colorectal 600c Rectum    NEc NEc 
AC Rectum 600 L2 spine, adj soft tissue    Pathologic PR PD 
AC Rectum 600 Posterior pelvis/sacrum    SD SD 
CholangioCA HCC Bile duct liver 600 Porta hepatis LN    SD PD 
AC Colon 900c RLQ abdomen    NEc NEc 
AC Ampulla 900 Liver Met    CR SD 
AC Rectum 900 Liver Met;    CR SD 
   R lung Met    PR  
AC Colon 900 Liver Met; R lung Met    SD SD 
AC Pancreas 1,200 L adrenal Met; para-aortic LN    SD SD 
SCC Esophagus 1,200 R supraclav LN    SD NE 
HCC Liver 1,200 R pulmonary artery Met; R lung hilum LN    PR PD 
AC Colon 1,800c Para-aortic, upper abd LN Dehydration NEc NEc 
    Pneumonia   
AC Bile duct 1,800 L upper tracheal supraclav LN Esophagitis/mucositis/anorexia/mouth pain SD PD 
    Dysphagia   
    Perianal irritation   
AC Colon 1,800c L acetabulum bone Met Anorexia NEc PD 
    Diarrhea   
AC Colon 1,200 Retroperit LN    SD SD 
SCC Esophagus 1,200c Gastrohep LN    NEc NEc 
Primary tumor histologyPrimary tumor siteIPdR dose (mg every day)Target site (RT field)Toxicity (≥grade 2 and attribution ≥ 3a)GradeAttribution to IPdRaTarget tumor responsebNontarget tumor responseb
AC Colon 150 Liver Met    SD SD 
AC Colon 300 Liver Met    SD SD 
AC Pancreas 600c Pancreas tail ALT PD at D 23c PD at D 23c 
    AST   
    rash   
AC Colorectal 600c Rectum    NEc NEc 
AC Rectum 600 L2 spine, adj soft tissue    Pathologic PR PD 
AC Rectum 600 Posterior pelvis/sacrum    SD SD 
CholangioCA HCC Bile duct liver 600 Porta hepatis LN    SD PD 
AC Colon 900c RLQ abdomen    NEc NEc 
AC Ampulla 900 Liver Met    CR SD 
AC Rectum 900 Liver Met;    CR SD 
   R lung Met    PR  
AC Colon 900 Liver Met; R lung Met    SD SD 
AC Pancreas 1,200 L adrenal Met; para-aortic LN    SD SD 
SCC Esophagus 1,200 R supraclav LN    SD NE 
HCC Liver 1,200 R pulmonary artery Met; R lung hilum LN    PR PD 
AC Colon 1,800c Para-aortic, upper abd LN Dehydration NEc NEc 
    Pneumonia   
AC Bile duct 1,800 L upper tracheal supraclav LN Esophagitis/mucositis/anorexia/mouth pain SD PD 
    Dysphagia   
    Perianal irritation   
AC Colon 1,800c L acetabulum bone Met Anorexia NEc PD 
    Diarrhea   
AC Colon 1,200 Retroperit LN    SD SD 
SCC Esophagus 1,200c Gastrohep LN    NEc NEc 

Abbreviations: L, left; LN, lymph nodes; Met, metastasis; R, right.

aAttribution: 1 = unrelated, 2 = unlikely, 3 = possible, 4 = probable, 5 = definite.

bRECIST criteria: NE, not evaluable.

cDid not complete study therapy.

In part I of the dose escalation scheme, the first and second patients received 150 and 300 mg orally every day, respectively, without DLT. The third study patient (600 mg orally every day) developed grade 3 transaminase elevation on Day 22 of study, in the context of presenting with fever and staphylococcus cellulitis at a previously removed mediport site (culture positive: staphylococcus). CT on Day 23 revealed an increase in the size and number of hepatic metastases (nontarget) and the size of the target lesion in the tail of the pancreas presumed to be progressive disease (PD), and the patient was removed from study on Day 24. At follow-up on Day 38, the cellulitis had resolved, transaminases were within normal limits, and no toxicities were noted. This event triggered the Stop/Switch rule, and part II of the dose escalation scheme was initiated.

Cohorts of 3 evaluable patients each for dose levels 600, 900, and 1,200 mg received therapy without evidence of IPdR-related grade 2 or higher clinical or laboratory toxicity. Four patients did not complete study therapy. One patient (1,200 mg) was removed from study on Day 2 per patient request. One patient (600 mg) developed PD after 22 days of IPdR and 12 days of RT, and discontinued all therapy. Two patients were removed from study before completion of IPdR therapy secondary to unrelated complications. One patient (600 mg) developed an incarceration of his colostomy on Day 9 (outside of RT field), and one patient (900 mg) developed a bowel obstruction from progressive colon cancer on Day 12 (outside of RT field). Both patients required emergent surgical intervention, and subsequently recovered. All patients were evaluable for toxicity for the duration of study therapy.

At 1,800 mg orally every day, 2 of 3 patients experienced grade 3 toxicity deemed probably or definitely related to IPdR. One patient required hospitalization for a community acquired pneumonia (CAP) on Day 7, with associated with cough and grade 3 dehydration. IPdR was possibly a factor in the development of the CAP and IPdR was considered to have contributed to the dehydration. The patient was removed from study (Day 10) with discontinuation of IPdR and interruption of RT (Day 3). The second patient with grade 3 toxicity at 1,800 mg every day presented on Day 22 of IPdR (Day 11 RT) with grade 3 diarrhea in the context of a right perianal abscess requiring incision and drainage. The patient was receiving RT to a left acetabular bone metastasis with a very limited RT field that did not include bowel or sphincter. The abscess was unrelated to study therapy, but it was felt that IPdR probably contributed to the patient's diarrhea. Of note in the context of documented infections, neither patient demonstrated leukopenia or neutropenia at any time. Both patients had resolution of their toxicity without sequelae. The MTD and RP2D based on this study is 1,200 mg orally every day.

Response

Although not the primary objective of the study, all patients had measurable disease at study entry. One patient developed PD of target and nontarget disease on Day 23 of study. Patients that completed study therapy had tumor assessment performed at week 8. Evaluation of target lesions using RECIST criteria revealed 2 complete response (CR), 3 partial response (PR), 9 stable disease (SD), and no PD in these patients (Table 2).

Study patients had metastatic disease and received no systemic therapy for at least 12 weeks (patients were required to be off therapy for at least 4 weeks prior to starting protocol and then received no systemic therapy for the 8 weeks of study therapy). Following assessment at week 8, many patients received additional systemic therapy, and thus, study-related response beyond week 8 could not be determined.

Pharmacokinetics

Our preclinical studies showed that the major determinant of IPdR-mediated tumor radiosensitization is the %IUdR-DNA cellular incorporation (40). As such, daily oral IPdR was started 1 week prior to the initiation of RT and continued throughout the RT course to maximize cumulative %IUdR-DNA tumor cell incorporation.

Plasma samples were analyzed for IPdR and its metabolites: IUdR, IUra, and IP. Specific pharmacologic information is not available for IPdR, IP, and IUra, but they were measured because their rates of metabolism influence total exposure to the active species, IUdR (Supplementary Fig. S1; Supplementary Materials and Methods).

Results of the pharmacokinetic analyses are illustrated in Figure 2. Figure 2A presents the mean values in each of 6 groups for plasma concentrations of IUdR (N = 18 patients total). As expected from our previously-published single-dose results (41), concentrations rose relatively quickly during the first hour, and persisted over the 4-hour sampling period. Concentrations increased consistently in each group as doses escalated. Longitudinal comparison across Days 1, 15, and 22 shows excellent consistency, especially for the higher doses that are most relevant to future planning.

Figure 2.

Pharmacokinetic analyses. A, Mean plasma concentrations of IUdR by IPdR dose level. B, Plasma concentrations of IUdR in individual patients on Days 1, 15, and 22 at 1,200 mg IPdR dose level (MTD; RP2D).

Figure 2.

Pharmacokinetic analyses. A, Mean plasma concentrations of IUdR by IPdR dose level. B, Plasma concentrations of IUdR in individual patients on Days 1, 15, and 22 at 1,200 mg IPdR dose level (MTD; RP2D).

Close modal

Figure 2B presents individual IUdR results for each of the 4 patients who received 1,200 mg, the MTD and RP2D. Interindividual consistency in concentrations was achieved among these patients within each day of sampling, and also across the days of sampling. Patient #14 was lower only on Day 15, and returned to highly consistent levels at Day 22. Patient #18 received 1,800 mg on Day 1, and then 1,200 mg every day Days 2 to 28.

RT is a mainstay of cancer therapy, with close to 60% of patients receiving RT at least once in the course of their treatment (1). RT is used as definitive treatment, in the neo-adjuvant and adjuvant settings to improve and consolidate results of surgical resection, and/or as palliation for cancer-related symptoms. RT can be delivered alone, or more commonly, in combination with chemotherapy and/or surgery. Finally, RT can be administered repeatedly in the course of therapy, either to new areas of disease or, in some cases, re-irradiation to a previously treated site.

The strategy to use a drug to selectively augment the therapeutic efficacy of RT is appealing. However, several issues specific to the mechanism and/or clinical use of radiosensitizers pose formidable challenges with regard to the development process for such agents. In the preclinical setting, the decision to pursue investigation of an oncologic agent is most often based on data from preclinical studies using models that rarely include RT. A selective radiosensitizer with limited or no direct cytotoxic activity fails to elicit responses in such models, and thus is likely to be abandoned. In the clinical arena, current standard-of-care cancer therapy involves multimodality treatment and multiagent chemotherapy that is often administered near the limits of normal tissue/organ tolerance. With potential toxicity and with limited direct antitumor activity, incorporating a radiosensitizer into these regimens is particularly challenging. Finally, pharmaceutical companies fear studying drugs in combination with RT, necessary for the investigation of a potential radiosensitizing agent, as any potential added toxicity could threaten established data regarding tolerability and use of their drug. The strategic guidelines collectively outlined by NCI, RTOG, and leading British investigators to promote the development of radiosensitizers address the distinct issues raised by the study of these agents and highlight the need to make such trials a priority (2, 3).

Several cytotoxic drugs are routinely administered with RT across a range of tumor types because the combined-modality approach has been shown to result in improved locoregional control, progression-free survival, or overall survival compared with single-modality treatment. The therapeutic improvements experienced with the addition of platins (e.g., cisplatin, carboplatin in head and neck, GI, cervix, and lung cancers; refs. 4–6), gemcitabine in pancreatic cancer (12), fluoropyrimidines (e.g., 5-FU, capecitabine in GI cancers; refs. 8–11), and alkylators (e.g., temozolomide in glioma; ref. 13) are welcome options, but they come at a cost of increased acute and late toxicities, thus limiting their therapeutic potential. Agents that target tumor-specific mediators of radiation response/repair would seem to be excellent candidates as radiosensitizing agents. To date, this approach has been largely unsuccessful, for example the inconsistent results and toxicities encountered with the use of EGFR inhibitors to block EGFR-related repair of radiation-induced DNA damage (42–44). How IPdR/IUdR compares in terms of radiosensitization and toxicity to other agents (5-FU and drugs that are not antimetabolites) will ultimately be determined by appropriately-designed clinical trials.

The toxicity profile of IPdR given with concurrent RT was notably favorable in this study. As is typical for phase I trials, the patients in this study were heavily pretreated, and eligibility required that patients have symptomatic metastatic disease necessitating palliative RT. DLT was encountered at 1,800 mg every day, when 2 of 3 patients experienced grade 3 toxicity. The grade 3 diarrhea experienced by 1 patient occurred in the context of an unrelated infection. The other patient developed CAP (with grade 3 dehydration) without evidence of immune compromise (i.e., normal peripheral blood counts). The manageable toxicity encountered in this study is particularly significant in light of the patients' clinical presentations (candidates for palliative RT) and the administration of concomitant, intermediate-dose RT.

The radiosensitization effects of IUdR have been appreciated for decades, but its clinical use is limited by the need for prolonged (24/7 for duration of RT course) infusion and moreover, by systemic toxicities at the doses required for radiosensitization. The extent of radiosensitization caused by IUdR is directly correlated with the amount of IUdR incorporated into tumor cell DNA, increasing with escalating plasma levels of IUdR without a known saturation point (18, 22, 45). In this trial, pharmacokinetic studies indicated that IPdR consistently produces plasma concentrations of the active metabolite, IUdR, within individual patients, and also across the patient population. The exact target range is not defined, but our previous work suggested concentrations of IUdR ≥1 μmol/L for 4 hours would mediate clinically relevant radiosensitization (17–20, 22–25). That goal was clearly met in this study. Results of this trial demonstrate the feasibility and tolerability of PO IPdR administered once daily in combination with an intermediate RT dose, and moreover, tolerable at doses that produce predictable and sustainable plasma IUdR levels that have been associated with clinically relevant radiosensitization.

Investigation of a potential radiosensitizing agent requires not only manipulation of the agent itself (i.e., dose, schedule), but also of the accompanying RT (i.e., dose, schedule, modality, and radiation source). In this study, an intermediate dose (37.5 Gy in 15 2.5 Gy fractions) was delivered by IMRT. Extrapolation of this data to other clinical scenarios, including different patient populations (e.g., tumor type, age, site, and extent of disease, etc.), additional concurrent treatment and alternative RT schemes must be performed with caution. If results of initial phase II studies are encouraging, separate clinical evaluation will be required to assess the toxicity and efficacy of IPdR as a radiosensitizing agent in a range of clinical situations.

The choice to target patients with GI malignancies in this trial was based in part on the expressed interest by the NCI Colorectal Cancer Working Group to investigate the addition of IPdR to standard-of-care therapy for patients with rectal cancer (11). The intermediate RT dose, schedule, and use of IMRT in this study provided results that can be applied to the development of follow-up clinical trials in patients with GI malignancies. Based on the results of this trial, a phase I/II trial of IPdR combining once daily IPdR with standard-of-care RT (45–50.4 Gy over 5–6 weeks) and twice daily capecitabine as neoadjuvant treatment of locally advanced (T3, T4, N1) rectal cancer using pathologic CR rate as the primary endpoint is under consideration.

In this trial, the administration of a single daily oral dose IPdR for 28 consecutive days with concurrent RT was shown to be feasible and tolerable at doses that produce reliable and sustainable plasma IUdR levels ≥1 μmol/L. Systemic toxicities were manageable, especially in light of the heavily pretreated patient population, indicating that the addition of IPdR to standard-of-care, established treatment regimens may be possible. If a phase II trial of IPdR + RT demonstrates a significant improvement in local tumor control with IPdR + RT, the impact could decrease rates of both local and distant failure, and perhaps permit lower RT dose for pediatric patients, elderly patients, and other patients for whom the toxicity of RT poses particular risk. Furthermore, the mechanism of IPdR/IUdR radiosensitization should theoretically extend to other forms of therapeutic RT including particle therapy and hypo- and hyperfractionation schedules. Thus, the results of this trial strongly support the investigation of the efficacy of IPdR + RT in phase II studies.

T. Kinsella and S. Wiersma are employees of and hold ownership interest (including patents) in EMEK, Inc. No potential conflicts of interest were disclosed by the other authors.

Conception and design: T. Kinsella, H. Safran, C. Kunos, J.M. Collins

Development of methodology: T. Kinsella, H. Safran, L.W. Anderson, K.D. Hill, C. Kunos, J.M. Collins

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T. Kinsella, T. DiPetrillo, A. Schumacher, J. Vatkevich, L.W. Anderson, K.D. Hill, C.A. Kunos

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T. Kinsella, H. Safran, T. DiPetrillo, L.W. Anderson, C. Kunos, J.M. Collins

Writing, review, and/or revision of the manuscript: T. Kinsella, H. Safran, S. Wiersma, T. DiPetrillo, C. Kunos, J.M. Collins

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Wiersma, A. Schumacher, J. Vatkevich, C. Kunos, J.M. Collins

Study supervision: T. Kinsella, H. Safran, A. Schumacher, J. Vatkevich, C. Kunos, J.M. Collins

Other (assisted the principal investigator with administrative support to document protocol amendments and memos): K. Rosati

This project has been funded in whole or in part with federal funds from the NCI, NIH, under contracts nos. HHSN261200800001E and HHSN261201400013C (Small Business Innovation Research contract through Shuttle Pharmaceuticals, Inc.). The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government. Support for this study was also provided by the DBJ Foundation and the University Radiation Medicine Foundation (to T. Kinsella).

BrUOG Co-investigators: R. Breakstone, K. Leonard, K. Mantripragada, M. LeGolvan

The patients who have participated in the clinical trials of IPdR.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
What is Radiation Therapy: American Society of Therapeutic Radiation Oncology (ASTRO)
; 
2018
.
Available from:
http://www.rtanswers.org/What-is-Radiation-Therapy.
2.
Lawrence
YR
,
Vikram
B
,
Dignam
JJ
,
Chakravarti
A
,
Machtay
M
,
Freidlin
B
, et al
NCI-RTOG translational program strategic guidelines for the early-stage development of radiosensitizers
.
J Nat Cancer Inst
2013
;
105
:
11
24
.
3.
Harrington
KJ
,
Billingham
LJ
,
Brunner
TB
,
Burnet
NG
,
Chan
CS
,
Hoskin
P
, et al
Guidelines for preclinical and early phase clinical assessment of novel radiosensitisers
.
Br J Cancer
2011
;
105
:
628
39
.
4.
Zuliani
AC
,
Esteves
SC
,
Teixeira
LC
,
Teixeira
JC
,
de Souza
GA
,
Sarian
LO
. 
Concomitant cisplatin plus radiotherapy and high-dose-rate brachytherapy versus radiotherapy alone for stage IIIB epidermoid cervical cancer: a randomized controlled trial
.
J Clin Oncol
2014
;
32
:
542
7
.
5.
Pignon
JP
,
le Maitre
A
,
Maillard
E
,
Bourhis
J
,
Group
M-NC
. 
Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): an update on 93 randomised trials and 17,346 patients
.
Radiother Oncol
2009
;
92
:
4
14
.
6.
Schaake-Koning
C
,
van den Bogaert
W
,
Dalesio
O
,
Festen
J
,
Hoogenhout
J
,
van Houtte
P
, et al
, 
Effects of concomitant cisplatin and radiotherapy on inoperable non-small-cell lung cancer
.
N Engl J Med
1992
;
326
:
524
30
.
7.
Bonner
JA
,
Harari
PM
,
Giralt
J
,
Azarnia
N
,
Shin
DM
,
Cohen
RB
, et al
Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck
.
N Engl J Med
2006
;
354
:
567
78
.
8.
Cooper
JS
,
Guo
MD
,
Herskovic
A
,
Macdonald
JS
,
Martenson
JA
 Jr.
,
Al-Sarraf
M
, et al
Chemoradiotherapy of locally advanced esophageal cancer: long-term follow-up of a prospective randomized trial (RTOG 85–01). Radiation Therapy Oncology Group
.
JAMA
1999
;
281
:
1623
7
.
9.
Orditura
M
,
Galizia
G
,
Sforza
V
,
Gambardella
V
,
Fabozzi
A
,
Laterza
MM
, et al
Treatment of gastric cancer
.
World J Gastroenterol
2014
;
20
:
1635
49
.
10.
Allegra
CJ
,
Yothers
G
,
O'Connell
MJ
,
Beart
RW
,
Wozniak
TF
,
Pitot
HC
, et al
Neoadjuvant 5-FU or capecitabine plus radiation with or without oxaliplatin in rectal cancer patients: a phase III randomized clinical trial
.
J Natl Cancer Inst
2015
;
107
(11):
djv248
.
11.
George
TJ
,
Franke
AJ
,
Chakravarthy
AB
,
Das
P
,
Dasari
A
,
El-Rayes
BF
, et al
National Cancer Institute (NCI) state of the science: Targeted radiosensitizers in colorectal cancer
.
Cancer
2019
;
125(16)
:
2732
46
.
12.
Yang
YF
,
Cao
XH
,
Bao
CE
,
Wan
X
. 
Concurrent radiotherapy with oral fluoropyrimidine versus gemcitabine in locally advanced pancreatic cancer: a systematic review and meta-analysis
.
Onco Targets Ther
2015
;
8
:
3315
22
.
13.
Stupp
R
,
Mason
WP
,
van den Bent
MJ
,
Weller
M
,
Fisher
B
,
Taphoorn
MJ
, et al
Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma
.
N Engl J Med
2005
;
352
:
987
96
.
14.
Ang
KK
. 
More lessons learned from the suffocation of hypoxia
.
J Clin Oncol
2010
;
28
:
2941
3
.
15.
Kinsella
TJ
. 
An approach to the radiosensitization of human tumors
.
Cancer J Sci Am
1996
;
2
:
184
93
.
16.
McGinn
CJ
,
Shewach
DS
,
Lawrence
TS
. 
Radiosensitizing nucleosides
.
J Nat Cancer Inst
1996
;
88
:
1193
203
.
17.
Rieber
M
,
Rieber
MS
. 
Sensitization to radiation-induced DNA damage accelerates loss of bcl-2 and increases apoptosis and autophagy
.
Cancer Biol Ther
2008
;
7
:
1561
6
.
18.
Kinsella
TJ
,
Dobson
PP
,
Mitchell
JB
,
Fornace
AJ
 Jr.
Enhancement of X ray induced DNA damage by pre-treatment with halogenated pyrimidine analogs
.
Int J Radiat Oncol Biol Phys
1987
;
13
:
733
9
.
19.
Lawrence
TS
,
Davis
MA
,
Maybaum
J
,
Stetson
PL
,
Ensminger
WD
. 
The effect of single versus double-strand substitution on halogenated pyrimidine-induced radiosensitization and DNA strand breakage in human tumor cells
.
Radiat Res
1990
;
123
:
192
8
.
20.
Fornace
AJ
 Jr.
,
Dobson
PP
,
Kinsella
TJ
. 
Enhancement of radiation damage in cellular DNA following unifilar substitution with iododeoxyuridine
.
Int J Radiat Oncol Biol Phys
1990
;
18
:
873
8
.
21.
Fowler
JF
,
Kinsella
TJ
. 
The limiting radiosensitization of tumors by S-phase sensitizers
.
Br J Cancer
1996
;
74
:
294
6
.
22.
Rodriguez
R
,
Ritter
MA
,
Fowler
JF
,
Kinsella
TJ
. 
Kinetics of cell labeling and thymidine replacement after continuous infusion of halogenated pyrimidines in vivo
.
Int J Radiat Oncol Biol Phys
1994
;
29
:
105
13
.
23.
Kinsella
TJ
,
Collins
J
,
Rowland
J
,
Klecker
R
 Jr.
,
Wright
D
,
Katz
D
, et al
Pharmacology and phase I/II study of continuous intravenous infusions of iododeoxyuridine and hyperfractionated radiotherapy in patients with glioblastoma multiforme
.
J Clin Oncol
1988
;
6
:
871
9
.
24.
Schulz
CA
,
Mehta
MP
,
Badie
B
,
McGinn
CJ
,
Robins
HI
,
Hayes
L
, et al
Continuous 28-day iododeoxyuridine infusion and hyperfractionated accelerated radiotherapy for malignant glioma: a phase I clinical study
.
Int J Radiat Oncol Biol Phys
2004
;
59
:
1107
15
.
25.
Urtasun
RC
,
Kinsella
TJ
,
Farnan
N
,
DelRowe
JD
,
Lester
SG
,
Fulton
DS
. 
Survival improvement in anaplastic astrocytoma, combining external radiation with halogenated pyrimidines: final report of RTOG 86–12, Phase I-II study
.
Int J Radiat Oncol Biol Phys
1996
;
36
:
1163
7
.
26.
Sullivan
FJ
,
Herscher
LL
,
Cook
JA
,
Smith
J
,
Steinberg
SM
,
Epstein
AH
, et al
National Cancer Institute (phase II) study of high-grade glioma treated with accelerated hyperfractionated radiation and iododeoxyuridine: results in anaplastic astrocytoma
.
Int J Radiat Oncol Biol Phys
1994
;
30
:
583
90
.
27.
Robertson
JM
,
Sondak
VK
,
Weiss
SA
,
Sussman
JJ
,
Chang
AE
,
Lawrence
TS
. 
Preoperative radiation therapy and iododeoxyuridine for large retroperitoneal sarcomas
.
Int J Radiat Oncol Biol Phys
1995
;
31
:
87
92
.
28.
Goffman
T
,
Tochner
Z
,
Glatstein
E
. 
Primary treatment of large and massive adult sarcomas with iododeoxyuridine and aggressive hyperfractionated irradiation
.
Cancer
1991
;
67
:
572
6
.
29.
Kinsella
TJ
,
Mitchell
JB
,
Russo
A
,
Morstyn
G
,
Glatstein
E
. 
The use of halogenated thymidine analogs as clinical radiosensitizers: rationale, current status, and future prospects: non-hypoxic cell sensitizers
.
Int J Radiat Oncol Biol Phys
1984
;
10
:
1399
406
.
30.
Speth
PA
,
Kinsella
TJ
,
Chang
AE
,
Klecker
RW
,
Belanger
K
,
Collins
JM
. 
Selective incorporation of iododeoxyuridine into DNA of hepatic metastases versus normal human liver
.
Clin Pharmacol Ther
1988
;
44
:
369
75
.
31.
Speth
PA
,
Kinsella
TJ
,
Chang
AE
,
Klecker
RW
 Jr.
,
Belanger
K
,
Smith
R
, et al
Iododeoxyuridine (IdUrd) incorporation into DNA of human hematopoietic cells, normal liver and hepatic metastases in man: as a radiosensitizer and as a marker for cell kinetic studies
.
Int J Radiat Oncol Biol Phys
1989
;
16
:
1247
50
.
32.
Epstein
AH
,
Lebovics
RS
,
Goffman
T
,
Teague
D
,
Fuetsch
ES
,
Glatstein
E
, et al
Treatment of locally advanced cancer of the head and neck with 5′-iododeoxyuridine and hyperfractionated radiation therapy: measurement of cell labeling and thymidine replacement
.
J Nat Cancer Inst
1994
;
86
:
1775
80
.
33.
Chang
CN
,
Doong
SL
,
Cheng
YC
. 
Conversion of 5-iodo-2-pyrimidinone-2′-deoxyribose to 5-iodo-deoxyuridine by aldehyde oxidase. Implication in hepatotropic drug design
.
Biochem Pharmacol
1992
;
43
:
2269
73
.
34.
Kinsella
TJ
,
Kunugi
KA
,
Vielhuber
KA
,
McCulloch
W
,
Liu
SH
,
Cheng
YC
. 
An in vivo comparison of oral 5-iodo-2′-deoxyuridine and 5-iodo-2-pyrimidinone-2′-deoxyribose toxicity, pharmacokinetics, and DNA incorporation in athymic mouse tissues and the human colon cancer xenograft, HCT-116
.
Cancer Res
1994
;
54
:
2695
700
.
35.
Kinsella
TJ
,
Vielhuber
KA
,
Kunugi
KA
,
Schupp
J
,
Davis
TW
,
Sands
H
. 
Preclinical toxicity and efficacy study of a 14-day schedule of oral 5-iodo-2-pyrimidinone-2′-deoxyribose as a prodrug for 5-iodo-2′-deoxyuridine radiosensitization in U251 human glioblastoma xenografts
.
Clin Cancer Res
2000
;
6
:
1468
75
.
36.
Kinsella
TJ
,
Kinsella
MT
,
Hong
S
,
Johnson
JP
,
Burback
B
,
Tosca
PJ
. 
Toxicology and pharmacokinetic study of orally administered 5-iodo-2-pyrimidinone-2′deoxyribose (IPdR) x 28 days in Fischer-344 rats: impact on the initial clinical phase I trial design of IPdR-mediated radiosensitization
.
Cancer Chemother Pharmacol
2008
;
61
:
323
34
.
37.
Kinsella
TJ
,
Kunugi
KA
,
Vielhuber
KA
,
Potter
DM
,
Fitzsimmons
ME
,
Collins
JM
. 
Preclinical evaluation of 5-iodo-2-pyrimidinone-2′-deoxyribose as a prodrug for 5-iodo-2′-deoxyuridine-mediated radiosensitization in mouse and human tissues
.
Clin Cancer Res
1998
;
4
:
99
109
.
38.
Kinsella
TJ
,
Schupp
JE
,
Davis
TW
,
Berry
SE
,
Hwang
HS
,
Warren
K
, et al
Preclinical study of the systemic toxicity and pharmacokinetics of 5-iodo-2-deoxypyrimidinone-2′-deoxyribose as a radiosensitizing prodrug in two, non-rodent animal species: implications for phase I study design
.
Clin Cancer Res
2000
;
6
:
3670
9
.
39.
Seo
Y
,
Yan
T
,
Schupp
JE
,
Colussi
V
,
Taylor
KL
,
Kinsella
TJ
. 
Differential radiosensitization in DNA mismatch repair-proficient and -deficient human colon cancer xenografts with 5-iodo-2-pyrimidinone-2′-deoxyribose
.
Clin Cancer Res
2004
;
10
:
7520
8
.
40.
Seo
Y
,
Yan
T
,
Schupp
JE
,
Radivoyevitch
T
,
Kinsella
TJ
. 
Schedule-dependent drug effects of oral 5-iodo-2-pyrimidinone-2′-deoxyribose as an in vivo radiosensitizer in U251 human glioblastoma xenografts
.
Clin Cancer Res
2005
;
11
:
7499
507
.
41.
Kummar
S
,
Anderson
L
,
Hill
K
,
Majerova
E
,
Allen
D
,
Horneffer
Y
, et al
First-in-human phase 0 trial of oral 5-iodo-2-pyrimidinone-2′-deoxyribose in patients with advanced malignancies
.
Clin Cancer Res
2013
;
19
:
1852
7
.
42.
Bonner
JA
,
Harari
PM
,
Giralt
J
,
Cohen
RB
,
Jones
CU
,
Sur
RK
, et al
Radiotherapy plus cetuximab for locoregionally advanced head and neck cancer: 5-year survival data from a phase 3 randomised trial, and relation between cetuximab-induced rash and survival
.
Lancet Oncol
2010
;
11
:
21
8
.
43.
Peereboom
DM
,
Shepard
DR
,
Ahluwalia
MS
,
Brewer
CJ
,
Agarwal
N
,
Stevens
GH
, et al
Phase II trial of erlotinib with temozolomide and radiation in patients with newly diagnosed glioblastoma multiforme
.
J Neurooncol
2010
;
98
:
93
9
.
44.
Govindan
R
,
Bogart
J
,
Stinchcombe
T
,
Wang
X
,
Hodgson
L
,
Kratzke
R
, et al
Randomized phase II study of pemetrexed, carboplatin, and thoracic radiation with or without cetuximab in patients with locally advanced unresectable non-small-cell lung cancer: Cancer and Leukemia Group B trial 30407
.
J Clin Oncol
2011
;
29
:
3120
5
.
45.
Miller
EM
,
Fowler
JF
,
Kinsella
TJ
. 
Linear-quadratic analysis of radiosensitization by halogenated pyrimidines. I. Radiosensitization of human colon cancer cells by iododeoxyuridine
.
Radiat Res
1992
;
131
:
81
9
.