Purpose: DMS612 is a dimethane sulfonate analog with bifunctional alkylating activity and preferential cytotoxicity to human renal cell carcinoma (RCC) in the NCI-60 cell panel. This first-in-human phase I study aimed to determine dose-limiting toxicity (DLT), maximum tolerated dose (MTD), pharmacokinetics (PK), and pharmacodynamics (PD) of DMS612 administered by 10-minute intravenous infusion on days 1, 8, and 15 of an every-28-day schedule.

Experimental Design: Patients with advanced solid malignancies were eligible. Enrollment followed a 3+3 design. PKs of DMS612 and metabolites were assessed by mass spectroscopy and PD by γ-H2AX immunofluorescence.

Results: A total of 31 patients, including those with colorectal (11), RCC (4), cervical (2), and urothelial (1) cancers, were enrolled. Six dose levels were studied, from 1.5 mg/m2 to 12 mg/m2. DLTs of grade 4 neutropenia and prolonged grade 3 thrombocytopenia were observed at 12 mg/m2. The MTD was determined to be 9 mg/m2 with a single DLT of grade 4 thrombocytopenia in 1 of 12 patients. Two patients had a confirmed partial response at the 9 mg/m2 dose level, in renal (1) and cervical (1) cancer. DMS612 was rapidly converted into active metabolites. γ-H2AX immunofluorescence revealed dose-dependent DNA damage in both peripheral blood lymphocytes and scalp hairs.

Conclusions: The MTD of DMS12 on days 1, 8, and 15 every 28 days was 9 mg/m2. DMS612 appears to be an alkylating agent with unique tissue specificities. Dose-dependent PD signals and two partial responses at the MTD support further evaluation of DMS612 in phase II trials. Clin Cancer Res; 21(4); 721–9. ©2014 AACR.

Translational Relevance

There is an unmet need for effective therapy against metastatic renal cell carcinoma (mRCC). Over the past decade, several agents targeting the VEGF and mTOR pathways have demonstrated clinical benefit. However, resistance develops in all cases, and median overall survival is still only about 2 years. For reasons that are poorly understood, cytotoxic chemotherapeutic agents have had limited efficacy in RCC. Using a high-throughput chemical library screen, we identified a group of dimethane sulfonates that were preferentially cytotoxic against RCC cell lines within the NCI-60 panel. These compounds are bifunctional alkylators that cluster together in COMPARE analysis and share a unique pattern of activity in the NCI Yeast Anticancer Drug Screen. The current report describes a first-in-human phase I clinical trial investigating DMS612, the lead compound of this group of dimethane sulfonates.

Benzaldehyde dimethane sulfonate (DMS612, NSC 281612, 4-[bis[2-[(methylsulfonyl)oxy]ethyl]amino]-2-methyl-benzaldehyde, BEN) was derived from a parent compound identified in the NCI-60 cell line Anticancer Drug Screen based on enhanced cytotoxicity against renal cell carcinoma (RCC; http://www.dtp.nci.nih.gov; ref. 1). The structure of DMS612 and preclinical evidence (2) suggests that this agent functions as a bifunctional alkylator with structural similarities to chlorambucil, busulfan, and melphalan (3). In contrast with these classical alkylating agents, DMS612 and related analogs were found to have additional activity against human RCC cell lines in the NCI-60 cell line screen (4). In vitro treatment of the human RCC cell line 109 and the breast cancer cell line MCF-7 with DMS612 caused cell-cycle arrest at the G2–M and S-phase (4) and resulted in increased p53 expression, indicating DNA damage. In the NCI Yeast Anticancer Drug Screen, DMS612 and related compounds were selectively toxic against yeast strains with mutations in rad18 (postreplication DNA repair), rad52 (recombination repair), and rad14 (nucleoside excision repair). Bioinformatic COMPARE analysis—using Pearson correlation of cell line GI50 values for a matrix of agents and cell lines found that DMS612 and related compounds reside in a unique cluster that is distinct from traditional alkylating agents, such as chlorambucil, carmustine, and busulfan (4). Given these characteristics, DMS612 was selected for further clinical evaluation.

Preclinical in vivo toxicology studies in rats and beagle dogs determined that dose-limiting toxicities (DLT) were mainly hematologic (leukopenia, thrombocytopenia, and reduced reticulocyte counts) and gastrointestinal (diarrhea and nausea/vomiting). The MTD of DMS612 dosed weekly three times was between 12 mg/m2/dose and 24 mg/m2/dose (2–4 mg/kg/dose) in Fischer 344 rats and greater than 30 mg/m2 in beagle dogs (1.5 mg/kg/dose).

DMS612 has demonstrated antitumor activity in in vivo xenograft models: DMS612 treatment in SCID female mice bearing human RCC RXF-393 xenografts (DMS612 Investigator Brochure), (5, 6) was able to produce tumor regressions at all doses and schedules studied; this antitumor activity was confirmed with additional xenograft models using orthotopic implantation of RCC lines ACHN-luc and 786-0, with greater activity seen against the latter (DMS612 Investigator Brochure).

In this first-in-human phase I study, we determined the DLT and MTD of DMS612 administered by 10-minute infusion on days 1, 8, and 15 of a 28-day cycle. We also characterized the pharmacokinetics (PK) of DMS612 and its active metabolites, and demonstrated pharmacodynamic (PD) evidence of induction of DNA damage response by quantification of γ-H2AX evolution using immunofluorescence in peripheral blood mononuclear cells (PBMC) and hair follicles at several time points during the first treatment.

Study design

This multicenter study (ClinicalTrials.gov Identifier: NCT00923520) was conducted at the NCI Clinical Center (Bethesda, MD), University of Pittsburgh Cancer Institute (Pittsburgh, PA), and Penn State Hershey Cancer Center (Hershey, PA), in accordance with the Declaration of Helsinki. The Institutional Review Boards at the respective institutions approved the study.

Patient selection

Eligible patients were ≥ 18 years of age with advanced solid tumors or lymphoma for which effective therapy did not exist or was no longer effective. There was no limit on prior chemotherapy treatment, although prior radiation to more than 25% of bone marrow was prohibited. Patients had to be ≥4 weeks from prior chemotherapy, monoclonal antibody therapy, or experimental therapy; ≥2 weeks from prior sorafenib, sunitinib, or temsirolimus treatment; or ≥6 weeks from prior mitomycin C or nitrosoureas. Patients were required to have acceptable organ and marrow function: leukocytes ≥3,000/μL, absolute neutrophil count ≥1,500/μL, platelets ≥100,000/μL total bilirubin within normal institutional limits, AST (SGOT) and ALT (SGPT) ≤2.5 X institutional upper limit of normal, creatinine within normal institutional limits, or creatinine clearance >50 mL/min per 1.73 m2 for patients with creatinine levels above institutional normal. The Eastern Cooperative Oncology Group performance status was required to be 0-2 and life expectancy ≥3 months. Toxicities from prior treatment must have resolved to ≤ grade 1 by Common Terminology Criteria for Adverse Events (CTCAE) v.4. Concomitant inhibitors and inducers of CYP3A4 were prohibited. Patients with central nervous system metastases were excluded, unless control had been achieved with either radiation or surgical resection at least 6 months before enrollment. Patients with uncontrolled medical illness including myocardial infarction within the past 6 months were excluded.

Study treatment and safety evaluation

DMS612 was supplied by the Division of Cancer Treatment and Diagnosis of the NCI in sterile, single-use vials containing a lyophilized powder/cake of 10 mg DMS612 and 1,000 mg hydroxypropyl-beta-cyclodextrin for reconstitution in 9.3 mL water for injection. After dilution in 0.9% sodium chloride or 5% dextrose, DMS612 was administered intravenously by central or peripheral catheter over 10 minutes on days 1, 8, and 15 of a 28-day cycle. There was no set limit on the number of cycles of study treatment administered. Dose escalation followed a modified Fibonacci schema starting at a dose of 1.5 mg/m2. A standard 3+3 design was used for dose escalation. Adverse events were assessed using CTCAE version 4.0. Vital signs and laboratory safety assessments were performed before each study treatment, and history and physical exam were performed at the start of each cycle.

Definition of DLT and MTD

DLT was defined as febrile neutropenia, or grade 4 neutropenia without fever for >5 days; grade 4 thrombocytopenia or anemia; grades ≥3 nonhematologic toxicity except for nausea, vomiting, diarrhea, or electrolyte abnormality corrected within 48 hours, or elevated creatinine corrected within 24 hours. The MTD was defined as the highest dose at which ≤1 of 6 patients experienced a DLT.

Response evaluation

Patients were evaluated for disease status by clinical and radiographic examination every two cycles (8 weeks) using RECIST 1.0 (7).

Pharmacokinetic analysis

Blood samples (4 mL) were collected in heparinized tubes on day 1 of the first cycle of therapy. Samples were collected before infusion, 5 minutes into infusion, and then at 2, 5, 15, 30, 45, 60, 120, 180, 240, 360, 480, and 1,440 minutes (24 hours) after the end of the 10-minute infusion. The samples were processed, and plasma concentrations of DMS612 and its metabolites were determined as previously described (2). PK parameters were extracted from the data by noncompartmental methods with PK Solutions 2.0 (Summit Research Services).

Pharmacodynamic assay of γ-H2AX detection in PBMCs

Blood was obtained before the first dose of DMS612 and at 120 and 240 minutes, and 24 hours following infusion of DMS612. Detection of γ-H2AX in PBMCs was performed as previously described (8). Briefly, PBMCs were isolated by Ficoll gradient, washed in 1× PBS, fixed in 2% paraformaldehyde (PFA), and spotted on slides by cytospin. PBMCs were permeabilized with prechilled ethanol 70%. Slides were stored at 4°C overnight then blocked for 30 minutes with 5% BSA before incubating 2 hours with a mouse monoclonal anti-γ-H2AX antibody (Millipore) and then 1 hour with a goat anti-mouse Alexa-488–conjugated IgG (Invitrogen). Finally, slides were incubated at 37°C for 5 minutes with a solution containing RNAse A (0.5 mg/mL) and propidium iodide (PI; 5 μg/mL). Slides were then mounted with mounting medium containing PI (Vectashield, Vector Laboratories, Inc.) and sealed with nail polish.

Pharmacodynamic assay of γ-H2AX detection in hair follicles and sample imaging

Hair samples were taken to measure γ-H2AX levels before infusion start and at approximately 120 and 240 minutes and 24 hours after the end of drug infusion on day 1 of the study.

Single hairs were plucked from the scalp with forceps (8). The objective was to acquire 12 hair that contained a fully intact follicle and sheath. Plucked hair bulbs were placed in 1× PBS on ice before fixation for 20 minutes at room temperature with 2% PFA. Following three washes in PBS, plucked hair was imaged and hair bulbs from anagen hair were selected for γ-H2AX detection. Samples were then permeabilized with prechilled ethanol 70%, washed, and attached to slides using a nail polish drop. γ-H2AX detection was performed as described above but incubation times with blocking and antibody solutions were increased by 50%. Washes were performed by slide immersion in PBS. Incubation with the RNAse (0.5 mg/mL) and PI (5 μg/mL) solution was performed at 37°C for 30 minutes and slides were mounted as described above. Hair shafts were then detached from the bulbs by using a razor blade and hair bulbs were mounted with mounting medium containing PI.

Samples were imaged by laser scanning confocal microscopy (Nikon PCM 2000, Nikon, Inc.). Optical sections through PBMCs and hair bulbs were combined in a maximum projection using the Simple 32 software (Compix Inc.). The foci were counted in PBMCs using the FociCounter software (9) by analyzing at least 200 cells, while foci were visually quantified by eye in the extremity of the hair bulbs.

Subject characteristics

Thirty-one patients with advanced solid tumors were enrolled in this phase I study of DMS612 administered on days 1, 8, and 15 of 28-day cycles. Patient characteristics are presented in Table 1. Patients with a range of solid tumors were enrolled, including 4 with RCC and 11 with colorectal carcinoma. The median number of prior chemotherapy regimens was 4 (range 0–13).

Table 1.

Subject characteristics

CharacteristicsNumber of subjects
Male 21 
Female 10 
Age (median, range) 59.4 y (41–75) 
Regimens prior chemotherapy (median, range) 4 (0–13) 
Prior surgery 24 
Prior radiation 11 
Prior immunotherapy 
Tumor type n 
 Chondrosarcoma 
 Carcinoid 
 Cervix 
 Cholangiocarinoma 
 Collecting duct of kidney 
 Colorectal 11 
 Gastroesophageal junction 
 Hepatocellular carcinoma 
 GIST 
 Mesothelioma (peritoneal) 
 Ovary 
 RCC 
 Small cell lung 
 Transitional cell carcinoma 
 Thymoma 
CharacteristicsNumber of subjects
Male 21 
Female 10 
Age (median, range) 59.4 y (41–75) 
Regimens prior chemotherapy (median, range) 4 (0–13) 
Prior surgery 24 
Prior radiation 11 
Prior immunotherapy 
Tumor type n 
 Chondrosarcoma 
 Carcinoid 
 Cervix 
 Cholangiocarinoma 
 Collecting duct of kidney 
 Colorectal 11 
 Gastroesophageal junction 
 Hepatocellular carcinoma 
 GIST 
 Mesothelioma (peritoneal) 
 Ovary 
 RCC 
 Small cell lung 
 Transitional cell carcinoma 
 Thymoma 

Study treatment and dose escalation

Patients were treated at six dose levels: 1.5 mg/m2 (n = 3), 3 mg/m2 (n = 3), 5 mg/m2 (n = 6), 7 mg/m2 (n = 3), 9 mg/m2 (n = 12), and 12 mg/m2 (n = 3). The number of cycles of study treatment ranged from 0 to 6 (median 2). The most common reason for discontinuation of study treatment was disease progression.

Toxicity

The DLTs observed in cycle 1 of study treatment are summarized in Table 2. Myelosuppression with thrombocytopenia and neutropenia was the principal DLT. Two of 3 patients treated at the 12 mg/m2 dose level experienced DLT: one developed grade 4 neutropenia and one had prolonged grade 3 thrombocytopenia. At the lower dose level of 9 mg/m2, only 1 of 12 patients experienced a DLT. As a result, the MTD was determined to be 9 mg/m2. Delayed and prolonged thrombocytopenia and neutropenia were observed in patients treated at the 9 and 12 mg/m2 dose levels, respectively. One of the patients with prolonged thrombocytopenia had metastatic RCC (mRCC) and had not received prior cytotoxic chemotherapy, suggesting that this toxicity may not be limited to patients with significant prior marrow exposure. Measured hematologic parameters eventually recovered in all of these patients after suspension of treatment. A bone marrow biopsy in one patient with prolonged thrombocytopenia showed normal trilineage hematopoiesis with decreased cellularity, consistent with the known effects on hematopoiesis seen more broadly with alkylating agents. Drug-related toxicities of all grades observed in cycle 1 of study treatment are presented in Table 3. Gastrointestinal toxicities were minimal and transient, and routine anti-emetic prophylaxis was not utilized. As shown in Table 3, grade 1 nausea was observed in 6 of 30 patients, and grade 2 nausea in only one patient.

Table 2.

Dose-limiting toxicities in cycle 1

Dose levelDose (mg/m2)Evaluated patients, nPrior chemotherapy regimens (median)DLT, nDLT
1.5 4.3 — 
2.3 — 
4.3 Infection Gr 5 
1.3 — 
12 3.4 PLT Gr 4 
12 ANC Gr 4 PLT prolonged Gr2/3 
Dose levelDose (mg/m2)Evaluated patients, nPrior chemotherapy regimens (median)DLT, nDLT
1.5 4.3 — 
2.3 — 
4.3 Infection Gr 5 
1.3 — 
12 3.4 PLT Gr 4 
12 ANC Gr 4 PLT prolonged Gr2/3 
Table 3.

Toxicities related to study treatment in cycle 1

Dose levelDL0 1.5 mg/m2 (N = 3)DL1 3 mg/m2 (N = 3)DL2 5 mg/m2 (N = 6)DL3 7 mg/m2 (N = 3)DL4 9 mg/m2 (N = 12)DL5 12 mg/m2 (N = 3)
Highest grade123412341234123412341234
Blood and lymphatic 
 Anemia                   
 Neutropenia                      1a 
 Leukopenia                   
 Lymphopenia                     
 Thrombocytopenia                 1a  1a  
Gastrointestinal 
 Diarrhea                      
 Nausea/vomiting                    
General 
 Anorexia                        
 Cardiac disorder, other                        
 Epistaxis                        
 Fatigue                    
 Fever                        
 Headache                        
 Infection            1a             
 Rash, acneform                        
 Thromboembolism                        
Laboratory/metabolic 
 Alk Phos                        
 AST                        
 Elevated Creatinine                        
 Hyperglycemia                      
 Hyperkalemia                        
 Hypokalemia                        
 Hypomagnesemia                        
 Hyponatremia                      
Dose levelDL0 1.5 mg/m2 (N = 3)DL1 3 mg/m2 (N = 3)DL2 5 mg/m2 (N = 6)DL3 7 mg/m2 (N = 3)DL4 9 mg/m2 (N = 12)DL5 12 mg/m2 (N = 3)
Highest grade123412341234123412341234
Blood and lymphatic 
 Anemia                   
 Neutropenia                      1a 
 Leukopenia                   
 Lymphopenia                     
 Thrombocytopenia                 1a  1a  
Gastrointestinal 
 Diarrhea                      
 Nausea/vomiting                    
General 
 Anorexia                        
 Cardiac disorder, other                        
 Epistaxis                        
 Fatigue                    
 Fever                        
 Headache                        
 Infection            1a             
 Rash, acneform                        
 Thromboembolism                        
Laboratory/metabolic 
 Alk Phos                        
 AST                        
 Elevated Creatinine                        
 Hyperglycemia                      
 Hyperkalemia                        
 Hypokalemia                        
 Hypomagnesemia                        
 Hyponatremia                      

aDLT.

Pharmacokinetics and metabolism

DMS612 was detected in only 2 patients (14.5 ng/mL midinfusion after 9 mg/m2; 4.7 ng/mL midinfusion after 12 mg/m2), and the half-life could not be determined in either of these patients. The PK parameters of a benzoic acid analog (BA) of DMS612, the primary enzymatic metabolite and putative alkylating agent; and BA-(OH)2, the major inactive, and ultimate metabolite in the detoxification pathway, are presented in the Supplementary Files, and a representative example is shown in Fig. 1A. Metabolic intermediates and glucuronide metabolites are shown in Supplementary Files (Supplementary Figs. S1A and S1B). The exposure (AUC0-t and Cmax) of BA (Fig. 1B) increased with dose. Exposure to BA appeared to increase more than linearly with dose as apparent clearance (Cl/F) of BA decreased with dose (Supplementary Fig. S1C), while the dose-normalized Cmax of BA increased with dose (Supplementary Fig. S1D).

Figure 1.

PK of DMS612 and metabolites. A, plasma concentration of BA (ng/mL) versus time for a patient who received 5.0 mg/m2 DMS612. BA () and BA-(OH)2 (). B, BA AUC versus dose. Patients experiencing DLT () and two patients with a response () are highlighted.

Figure 1.

PK of DMS612 and metabolites. A, plasma concentration of BA (ng/mL) versus time for a patient who received 5.0 mg/m2 DMS612. BA () and BA-(OH)2 (). B, BA AUC versus dose. Patients experiencing DLT () and two patients with a response () are highlighted.

Close modal

Pharmacodynamics

An antibody-based immunofluorescence assay to detect the phosphorylated form of the histone variant H2AX referred to as γ-H2AX, was used in this study to evaluate DNA damage response in surrogate tissues during cycle 1 of treatment. Two different tissues were evaluated in eligible patients: PBMCs and plucked hair bulbs. These were examined for DMS612-induced DNA damage (Fig. 2), as indicated by the induction of γ-H2AX foci. At the lowest dose, 1.5 mg/m2, no γ-H2AX foci formed at any time up to 24 hours. At 3 and 5 mg/m2, some induction of γ-H2AX foci is apparent in PBMCs at 24 hours, while at 7, 9 and 12 mg/m2, induction is apparent at 4 hours. γ-H2AX immunofluorescence at 24 hours was dose dependent, with the 9 and 12 mg/m2 values being about 3-fold higher than in the lower doses.

Figure 2.

Pharmacodynamic assessment of γ-H2AX formation following DMS612 study treatment. γ-H2AX formation in PBMCs and plucked hair bulbs from patients receiving escalating doses of DMS612 (1.5, 3, 5, 7, 9, and 12 mg/m2). Blood and hair samples were collected before (Pre) and 2, 4, and 24 hours after infusions of DMS612. A, γ-H2AX in PBMCs. Left, data from individuals plotted as percentage of positive cells (more than four foci per cell; fpc). Right, data plotted as positive cells ± SDs (n = 4 individuals for 1.5 mg/m2, n = 2 for 3 mg/m2, n = 2 for 5 mg/m2, n = 2 for 7 mg/m2, n = 5 for 9 mg/m2, and n = 1 for 12 mg/m2). Values for γ-H2AX foci per cell are presented in Supplementary Table S2. B, γ-H2AX formation in plucked hair bulbs. Left, data from individuals plotted as percentage of positive cells (more than four fpc). Right, data plotted as positive cells ± SDs (n = 2 individuals for 1.5 mg/m2, n = 2 for 3 mg/m2 and n = 3 for 9 mg/m2). The colored rectangles in the left panels of A and B refer to blood and hair samples obtained from the same patients. Details of the γ-H2AX analysis for plucked hairs are presented in Supplemental Table S3. C, representative images of γ-H2AX staining in patients' PBMCs and plucked hair bulbs collected before and during DMS612 infusion of 9 mg/m2 (PBMCs, left) and 3 mg/m2 (plucked hairs, right). Green, γ-H2AX; red, DNA stained with PI. Insets (high magnification images) in the right panel show more clearly DNA damage (green) in hair nuclei (red). The image on the left of the right panel shows regions of plucked hair bulbs where both image capture and γ-H2AX quantification were performed (white box).

Figure 2.

Pharmacodynamic assessment of γ-H2AX formation following DMS612 study treatment. γ-H2AX formation in PBMCs and plucked hair bulbs from patients receiving escalating doses of DMS612 (1.5, 3, 5, 7, 9, and 12 mg/m2). Blood and hair samples were collected before (Pre) and 2, 4, and 24 hours after infusions of DMS612. A, γ-H2AX in PBMCs. Left, data from individuals plotted as percentage of positive cells (more than four foci per cell; fpc). Right, data plotted as positive cells ± SDs (n = 4 individuals for 1.5 mg/m2, n = 2 for 3 mg/m2, n = 2 for 5 mg/m2, n = 2 for 7 mg/m2, n = 5 for 9 mg/m2, and n = 1 for 12 mg/m2). Values for γ-H2AX foci per cell are presented in Supplementary Table S2. B, γ-H2AX formation in plucked hair bulbs. Left, data from individuals plotted as percentage of positive cells (more than four fpc). Right, data plotted as positive cells ± SDs (n = 2 individuals for 1.5 mg/m2, n = 2 for 3 mg/m2 and n = 3 for 9 mg/m2). The colored rectangles in the left panels of A and B refer to blood and hair samples obtained from the same patients. Details of the γ-H2AX analysis for plucked hairs are presented in Supplemental Table S3. C, representative images of γ-H2AX staining in patients' PBMCs and plucked hair bulbs collected before and during DMS612 infusion of 9 mg/m2 (PBMCs, left) and 3 mg/m2 (plucked hairs, right). Green, γ-H2AX; red, DNA stained with PI. Insets (high magnification images) in the right panel show more clearly DNA damage (green) in hair nuclei (red). The image on the left of the right panel shows regions of plucked hair bulbs where both image capture and γ-H2AX quantification were performed (white box).

Close modal

Increases in γ-H2AX immunofluorescence were more pronounced in plucked hair bulbs than in PBMCs, appearing earlier and with more numerous foci (Fig. 2). For example, in PBMCs, the signal at 3 mg/m2 was about 25% of that at 9 mg/m2 after 24 hours, whereas in the hair bulbs, the γ-H2AX apparent at 3 mg/m2 was 65% of that at 9 mg/m2. Interestingly, DNA damage, as reflected by expression of the γ-H2AX biomarker, appeared to correlate with the incidence and severity of myelosuppression, supporting the hypothesis that DNA damage is biologically relevant to the mechanisms of action of DMS612.

Tumor responses

One patient with RCC and one patient with cervical cancer had a confirmed partial response. Both patients were treated at the MTD of 9 mg/m2 (Fig. 3). Stable disease of at least 16-week duration was observed in 3 additional patients [colon (2) and RCC (1)].

Figure 3.

Tumor responses during DMS612 study treatment. A, effect of DMS on metastatic cervical carcinoma following three cycles of treatment, achieving confirmed partial response (only day 1 treatment administered in cycle 3 due to G3 leukopenia and G3 neutropenia). CT images obtained at 5-mm cuts show disappearance of a pericardial nodule. The apparent difference in level of cut is due to a different breathhold and 5-mm cut versus 2-mm cut at baseline. B, additional sites including pathologically enlarged axillary lymphadenopathy also demonstrated response. C, effect of DMS on patient with mRCC with partial response confirmed, including gastric lymph node (left) and right anterior chest wall mass involving destruction of the acromial extremity of the right clavicle (right).

Figure 3.

Tumor responses during DMS612 study treatment. A, effect of DMS on metastatic cervical carcinoma following three cycles of treatment, achieving confirmed partial response (only day 1 treatment administered in cycle 3 due to G3 leukopenia and G3 neutropenia). CT images obtained at 5-mm cuts show disappearance of a pericardial nodule. The apparent difference in level of cut is due to a different breathhold and 5-mm cut versus 2-mm cut at baseline. B, additional sites including pathologically enlarged axillary lymphadenopathy also demonstrated response. C, effect of DMS on patient with mRCC with partial response confirmed, including gastric lymph node (left) and right anterior chest wall mass involving destruction of the acromial extremity of the right clavicle (right).

Close modal

This is a first-in-human phase I study to evaluate the safety, DLT, MTD, and PK and PD of DMS612 administered weekly for 3 of every 4 weeks. In patients with previously treated solid tumors, the MTD of DMS612 was 9 mg/m2 when administered on days 1, 8, and 15 of a 28-day cycle. DLTs of neutropenia and thrombocytopenia were consistent with the proposed mechanism of DMS612 as an alkylating agent. In some patients, myelosuppression was prolonged, but recovery of measured hematologic parameters was documented in every subject. While reversible myelosuppression was observed, toxicities such as alopecia and peripheral neuropathy were not, and gastrointestinal toxicity was relatively modest. When combined with the in vitro observations regarding the activity of DMS612 in renal cell cancer cell lines, these findings suggest that DMS612 is an alkylating agent with unique tissue specificities that remain presently unexplained and, therefore, make the agent an interesting and unique compound for further study

PK analysis revealed that DMS612 was rapidly metabolized as parent drug was detected in only 2 patients at the earliest time point: 2 minutes after completion of infusion. This result is consistent with preclinical murine studies in which DMS612 was detected only within the first 10 minutes of an intravenous bolus administration of drug, confirming the extremely rapid metabolism of DMS612 to BA. The BA Cmax in humans of approximately 4,000 ng/mL after 12 mg/m2 DMS612 is in good agreement with the Cmax of 18,000 ng/mL observed in mice after 60 mg/m2 (20 mg/kg; ref. 3), arguing for the validity of the murine model for DMS612 pharmacology. PK analysis identified a large interpatient variability in the concentration and AUC of BA, the major metabolite of DMS612 (2). BA was present in all but 2 patients; however, these patients did have downstream BA metabolites detected. The BA plasma half-life was several-fold longer than the 5 minute in vitro plasma half-life of this highly reactive compound, suggesting continued metabolic generation of BA from DMS612 in tissues, followed by redistribution into the plasma compartment. This finding is indirect evidence of the ability of DMS612 to reach tissues with subsequent intracellular activation to BA, the likely effector of DNA damage, as previously suggested (2). These observations are substantiated by the findings from γ-H2AX assays, which show that DMS612 leads to DNA damage induction in both blood cells and hair follicles. The capacity for DMS612 to generate DNA breaks in complex tissue (i.e., hair follicles) suggests that the drug most likely reaches and induces damage in solid tumors. The selective cytotoxicity of the benzaldehyde dimethane sulfonate compounds to yeast strains with specific defects in DNA repair suggests that the status of DNA repair pathways in tumor cells is critical for determining DMS612 activity (4).

All of the DMS612 metabolites and respective glucuronide forms that were detected in preclinical studies were also observed in patients, supporting the translation of previous preclinical findings related to DMS612 pharmacology. DMS612 metabolism differs from that of conventional alkylating agents in a number of ways. DMS612 is a lipophilic prodrug of BA; intracellularly generated, BA is a much more reactive analog and the likely alkylator of cellular targets. We previously demonstrated that aldehyde dehydrogenase (ALDH) may be responsible for the intracellular metabolism of DMS612 to BA, and that DMS612 is more active in cell lines that express high levels of ALDH (ref. 2; and manuscript in preparation). Therefore, DMS612 appears to have unique characteristics that are quite different from other well-characterized alkylating agents. The importance may lie in the reported overexpression of ALDH in stem cells of many tumor types making these stem cells potential targets for DMS612 therapy (10). The prolonged myelosuppression observed with DMS612 is suggestive of enhanced toxicity to bone marrow stem cells. Intriguingly, hematopoietic stem cells express high levels of ALDH (11), potentially facilitating the rapid conversion of DMS612 to BA, resulting in higher levels of intracellular alkylation and cytotoxicity. The short half-life of DMS612 might be a favorable property for use in antibody–drug conjugates, or in nanoparticle drug delivery systems. Such an approach might also circumvent some of the bone marrow toxicity.

One patient with prolonged myelosuppression underwent evaluation with bone marrow aspiration and biopsy, which revealed marked hypocellularity but with normal trilineage hematopoiesis, a finding consistent with the known effects of other well-established alkylating agents. An alternative schedule of drug administration on days 1 and 2 on an every 21-day schedule is currently being investigated in a phase I cohort of patients with advanced solid tumors. We hypothesize that the prolonged (20-day) interval of this modified schedule would allow for greater hematopoietic recovery, and would potentially be associated with reduced myelosuppression at clinically effective doses. The patient population that received DMS612 includes patients with multiple prior therapies, median 4 (range 0 – 13). Cumulative myelosuppression is more likely in patients with extensive prior cytotoxic therapies. As a result, patients enrolled on this study may not predict tolerability in patients with RCC, who are principally treated with targeted agents and immunotherapy that is not generally associated with severe or prolonged myelosuppression.

In the study presented here, tumor responses were observed in one patient with mRCC and in one patient with advanced cervical carcinoma. The fact that these responses were observed in a heavily pretreated population with a first-in-human agent is certainly interesting, and warrants further investigation of antitumor activity. RCC is generally not responsive to traditional cytotoxic agents used to treat other solid tumors such as alkylating agents, plant alkaloids including the taxanes, and antimetabolites (12, 13). At present, the mechanisms of RCC chemoresistance are not well understood. However, contributing factors include overexpression of several important proteins, including multidrug resistance proteins, major vault protein, and glutathione-s-transferase (12, 14–16). The degree to which DMS612 is a substrate of these proteins is unknown. Notably, the sensitivity pattern for an ABCB1 substrate in the NCI-60 was not observed for DMS612 (1). The dimethane sulfonate compounds shared a unique profile of specificity against yeast strains with particular DNA repair mutations (4). This pattern of sensitivity could potentially be exploited for identification of a biomarker for predicting response among individual tumors with specific defects in DNA repair (17). Another potential biomarker is ATAD5, a DNA repair protein with polymorphic variants that were identified as highly correlated with DMS612 sensitivity in the NCI 60 cell line drug screen (18). Elucidating which DNA repair proteins are important for DMS612 sensitivity and resistance could allow patient selection or stratification in future clinical trials.

In conclusion, the predictable and well-tolerated safety profile and preliminary antitumor activity supported by pharmacodynamic evidence of DNA damage suggest that DMS612 is suitable for phase II studies in RCC and other solid tumors. The recent approvals of pemetrexed for non–small cell lung cancer and bendamustine for chronic lymphocytic leukemia and indolent B-cell lymphoma demonstrate that novel cytotoxic agents will continue to provide advances in cancer treatment even in this era of targeted therapy. DMS612 offers the interesting possibility of an alkylating agent with a unique toxicity profile. The lack of alopecia, nausea, or mucositis suggests the drug may not be activated in all tissues, and means improved tolerability over alkylating agents of the past. Strategies to avoid the cumulative myelotoxicity and to exploit DMS612 in renal cancer should be vigorously pursued.

No potential conflicts of interest were disclosed.

Conception and design: L.J. Appleman, S. Balasubramaniam, J.J. Wright, D.R. Kohler, C.P. Belani, J.H. Beumer, S.E. Bates

Development of methodology: S. Balasubramaniam, R.A. Parise, C.E. Redon, D.R. Kohler, C.P. Belani, J. Eiseman, J.H. Beumer, S.E. Bates

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L.J. Appleman, S. Balasubramaniam, R.A. Parise, C. Bryla, C.E. Redon, A.J. Nakamura, W.M. Bonner, D.R. Kohler, Y. Jiang, C.P. Belani, J. Eiseman, J.H. Beumer, S.E. Bates

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L.J. Appleman, S. Balasubramaniam, R.A. Parise, C. Bryla, C.E. Redon, W.M. Bonner, C.P. Belani, E. Chu, J.H. Beumer, S.E. Bates

Writing, review, and/or revision of the manuscript: L.J. Appleman, S. Balasubramaniam, R.A. Parise, C. Bryla, C.E. Redon, W.M. Bonner, J.J. Wright, R. Piekarz, D.R. Kohler, C.P. Belani, J. Eiseman, E. Chu, J.H. Beumer, S.E. Bates

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Balasubramaniam, C. Bryla, R. Piekarz, D.R. Kohler, J.H. Beumer

Study supervision: S. Balasubramaniam, C. Bryla, J.J. Wright, J.H. Beumer, S.E. Bates

The authors thank the Merrill Egorin Writing Group at UPCI for their constructive comments and input regarding the manuscript, the late Dr. Merrill Egorin for his input in the original design of this trial, the clinical trial group of Dr. Tito Fojo for participation in the conduct of this trial, and Rob Robey for editorial assistance.

This work was supported, in part, by the National Cancer Institute grants U01-CA099168 (to L.J. Appleman, R.A. Parise, Y. Jiang, C.P. Belani, J. Eiseman, E. Chu, and J.H. Beumer) and P30-CA47904 (to J.H. Beumer and R.A. Parise), and by the Intramural Research Program of the NIH, National Cancer Institute (to S. Balasubramaniam, C. Bryla, W.M. Bonner, C.E. Redon, A.J. Nakamura, J.J. Wright, R.A. Parise, and S.E. Bates).

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.
Mertins
SD
,
Myers
TG
,
Hollingshead
M
,
Dykes
D
,
Bodde
E
,
Tsai
P
, et al
Screening for and identification of novel agents directed at renal cell carcinoma
.
Clin Cancer Res
2001
;
7
:
620
33
.
2.
Parise
RA
,
Anyang
BN
,
Eiseman
JL
,
Egorin
MJ
,
Covey
JM
,
Beumer
JH
. 
Formation of active products of benzaldehyde dimethane sulfonate (NSC 281612, DMS612) in human blood and plasma and their activity against renal cell carcinoma lines
.
Cancer Chemother Pharmacol
2013
;
71
:
73
83
.
3.
Parise
RA
,
Beumer
JH
,
Clausen
DM
,
Rigatti
LH
,
Ziegler
JA
,
Gasparetto
M
, et al
Effects of the aldehyde dehydrogenase inhibitor disulfiram on the plasma pharmacokinetics, metabolism, and toxicity of benzaldehyde dimethane sulfonate (NSC281612, DMS612, BEN) in mice
.
Cancer Chemother Pharmacol
2013
;
72
:
1195
204
.
4.
Mertins
SD
,
Myers
TG
,
Holbeck
SL
,
Medina-Perez
W
,
Wang
E
,
Kohlhagen
G
, et al
In vitro evaluation of dimethane sulfonate analogues with potential alkylating activity and selective renal cell carcinoma cytotoxicity
.
Mol Cancer Ther
2004
;
3
:
849
60
.
5.
Mertins
S
. 
Treating renal cancer using a 4-[Bis[2-[(methylsulfonyl)oxy]ethyl]amino]-2-methyl-benzaldehyde
.
United States
:
Patent Application Publication
; 
2011
.
6.
Carter
J
. 
In vivo efficacy of an aldehyde degradation product of dimethane sulfonate (NSC 281612) in an orthotopic RXF-393 human renal tumor model
.
Proc Am Assoc Cancer Res
2005
;
2005
;
322
3
.
7.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
8.
Redon
CE
,
Nakamura
AJ
,
Sordet
O
,
Dickey
JS
,
Gouliaeva
K
,
Tabb
B
, et al
gamma-H2AX detection in peripheral blood lymphocytes, splenocytes, bone marrow, xenografts, and skin
.
Methods Mol Biol
2011
;
682
:
249
70
.
9.
Jucha
A
,
Wegierek-Ciuk
A
,
Koza
Z
,
Lisowska
H
,
Wojcik
A
,
Wojewodzka
M
, et al
FociCounter: a freely available PC programme for quantitative and qualitative analysis of gamma-H2AX foci
.
Mutat Res
2010
;
696
:
16
20
.
10.
Allahverdiyev
AM
,
Bagirova
M
,
Oztel
ON
,
Yaman
S
,
Abamor
ES
,
Koc
RC
, et al
In:
Canuto
RA
,
editor
. 
Aldehyde dehydrogenase
:
Cancer and stem cells
; 
2012
.
Available from
: http://www.intechopen.com/books/dehydrogenases/aldehyde-dehydrogenase-cancer-and-stem-cells.
11.
Kastan
MB
,
Schlaffer
E
,
Russo
JE
,
Colvin
OM
,
Civin
CI
,
Hilton
J
. 
Direct demonstration of elevated aldehyde dehydrogenase in human hematopoietic progenitor cells
.
Blood
1990
;
75
:
1947
50
.
12.
Gamelin
E
,
Mertins
SD
,
Regis
JT
,
Mickley
L
,
Abati
A
,
Worrell
RA
, et al
Intrinsic drug resistance in primary and metastatic renal cell carcinoma
.
J Urol
1999
;
162
:
217
24
.
13.
Yagoda
A
,
Abi-Rached
B
,
Petrylak
D
. 
Chemotherapy for advanced renal-cell carcinoma: 1983–1993
.
Semin Oncol
1995
;
22
:
42
60
.
14.
Walsh
N
,
Larkin
A
,
Kennedy
S
,
Connolly
L
,
Ballot
J
,
Ooi
W
, et al
Expression of multidrug resistance markers ABCB1 (MDR-1/P-gp) and ABCC1 (MRP-1) in renal cell carcinoma
.
BMC Urol
2009
;
9
:
6
.
15.
Ferguson
RE
,
Jackson
SM
,
Stanley
AJ
,
Joyce
AD
,
Harnden
P
,
Morrison
EE
, et al
Intrinsic chemotherapy resistance to the tubulin-binding antimitotic agents in renal cell carcinoma
.
Int J Cancer
2005
;
115
:
155
63
.
16.
Chuang
ST
,
Chu
P
,
Sugimura
J
,
Tretiakova
MS
,
Papavero
V
,
Wang
K
, et al
Overexpression of glutathione s-transferase alpha in clear cell renal cell carcinoma
.
Am J Clin Pathol
2005
;
123
:
421
9
.
17.
Postel-Vinay
S
,
Vanhecke
E
,
Olaussen
KA
,
Lord
CJ
,
Ashworth
A
,
Soria
JC
. 
The potential of exploiting DNA-repair defects for optimizing lung cancer treatment
.
Nat Rev Clin Oncol
2012
;
9
:
144
55
.
18.
Abaan
OD
,
Polley
EC
,
Davis
SR
,
Zhu
YJ
,
Bilke
S
,
Walker
RL
, et al
The exomes of the NCI-60 panel: a genomic resource for cancer biology and systems pharmacology
.
Cancer Res
2013
;
73
:
4372
82
.