+ All Categories
Home > Documents > Talabostat

Talabostat

Date post: 14-Dec-2016
Category:
Upload: charles-casey
View: 219 times
Download: 1 times
Share this document with a friend
8
Drug Evaluation 10.1517/13543784.16.9.1459 © 2007 Informa UK Ltd ISSN 1354-3784 1459 1. Introduction 2. Mechanism of action 3. Preclinical studies 4. Clinical studies 5. Conclusions 6. Expert opinion Talabostat Charles Casey Cunningham Mary Crowley Medical Research Center, 1717 Main Street, 60th Floor, Dallas, Texas, 75201, USA Talabostat mesilate is an orally active, specific inhibitor of dipeptidyl peptidases, including tumor-associated fibroblast activation protein. However, by an independent mechanism, talabostat also stimulates the upregulation of cytokines and chemokines to engender a tumor-specific host immune response, thus giving it a unique dual mechanism of action. In clinical trials, talabostat has demonstrated significant activity, including achieving complete responses in patients with non-small-cell lung cancer and malignant melanoma. Keywords: CD26/dipeptidyl peptidase IV , fibroblast activation protein, metastatic melanoma, non-Hodgkin ’ s lymphoma, non-small cell lung cancer , talabostat, tumor stroma Expert Opin. Investig. Drugs (2007) 16(9):1459-1465 1. Introduction Talabostat mesilate (PT-100) is the methanesulfonate salt of L-valinyl- L-boroproline (chemical structure shown in Figure 1) [1]. Amino boronic dipeptides are attractive as potential therapeutics because of their high affinity for the catalytic site of several serine proteases [2]. Of these, talabostat is the most extensively developed clinically. Talabostat is orally available and specifically inhibits dipeptidyl peptidases such as fibroblast activation protein (FAP) [3] and dipeptidyl peptidase IV (DPP-IV)/CD26 [4,5]. 2. Mechanism of action Although developed as an inhibitor of dipeptidyl peptidases, talabostat also causes a variety of immune effects in animal models by increasing the production of cytokine and chemokines in both lymphoid organs and tumor mass, leading to enhancement of tumor-specific T-cell immunity [6] as well as T-cell-independent stimulation of the antitumor activity of neutrophils, macrophages and natural killer cells [7,8]. The precise mechanism by which these effects occur is not fully known, but talabostat does activate caspase-1 in preclinical models, thereby inducing an IL-1 β response in tumor stroma and associated lymph nodes (B Jones, Point Therapeutics, pers. commun.). Therefore, cytokine production would occur predominantly within the tumor tissue itself, thus encouraging a local and tumor-specific response. Notably, this effect is enhanced by the addition of any of several chemotherapy agents, including cisplatin, gemcitabine, paclitaxel and 5-fluorouracil, as well as the monoclonal antibody rituximab in the case of the lymphoma model. One possibility is that chemotherapy-induced apoptosis leads to exposure of tumor- associated antigens and thus promotes antigen processing. Subsequent treatment with talabostat might then promote the maturation of professional antigen- presenting cells and the stimulation of antigen-specific T cells, including cytolytic T lymphocytes, via the upregulation of cytokine and chemokine production within the tumor and lymphoid tissues. 3. Preclinical studies In preclinical studies, talabostat demonstrates significant antitumor effects, both as a single agent and in combination with chemotherapy or monoclonal antibodies [6-8]. Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by University of Connecticut on 12/04/13 For personal use only.
Transcript
Page 1: Talabostat

Drug Evaluation

10.1517/13543784.16.9.1459 © 2007 Informa UK Ltd ISSN 1354-3784 1459

1. Introduction

2. Mechanism of action

3. Preclinical studies

4. Clinical studies

5. Conclusions

6. Expert opinion

Talabostat Charles Casey Cunningham Mary Crowley Medical Research Center, 1717 Main Street, 60th Floor, Dallas, Texas, 75201, USA

Talabostat mesilate is an orally active, specific inhibitor of dipeptidyl peptidases, including tumor-associated fibroblast activation protein. However, by an independent mechanism, talabostat also stimulates the upregulation of cytokines and chemokines to engender a tumor-specific host immune response, thus giving it a unique dual mechanism of action. In clinical trials, talabostat has demonstrated significant activity, including achieving complete responses in patients with non-small-cell lung cancer and malignant melanoma.

Keywords: CD26/dipeptidyl peptidase IV , fibroblast activation protein , metastatic melanoma , non-Hodgkin ’ s lymphoma , non-small cell lung cancer , talabostat , tumor stroma

Expert Opin. Investig. Drugs (2007) 16(9):1459-1465

1. Introduction

Talabostat mesilate (PT-100) is the methanesulfonate salt of L -valinyl- L -boroproline (chemical structure shown in Figure 1 ) [1] . Amino boronic dipeptides are attractive as potential therapeutics because of their high affinity for the catalytic site of several serine proteases [2] . Of these, talabostat is the most extensively developed clinically. Talabostat is orally available and specifically inhibits dipeptidyl peptidases such as fibroblast activation protein (FAP) [3] and dipeptidyl peptidase IV (DPP-IV)/CD26 [4,5] .

2. Mechanism of action

Although developed as an inhibitor of dipeptidyl peptidases, talabostat also causes a variety of immune effects in animal models by increasing the production of cytokine and chemokines in both lymphoid organs and tumor mass, leading to enhancement of tumor-specific T-cell immunity [6] as well as T-cell-independent stimulation of the antitumor activity of neutrophils, macrophages and natural killer cells [7,8] . The precise mechanism by which these effects occur is not fully known, but talabostat does activate caspase-1 in preclinical models, thereby inducing an IL-1 β response in tumor stroma and associated lymph nodes (B Jones, Point Therapeutics, pers. commun.). Therefore, cytokine production would occur predominantly within the tumor tissue itself, thus encouraging a local and tumor-specific response.

Notably, this effect is enhanced by the addition of any of several chemotherapy agents, including cisplatin, gemcitabine, paclitaxel and 5-fluorouracil, as well as the monoclonal antibody rituximab in the case of the lymphoma model. One possibility is that chemotherapy-induced apoptosis leads to exposure of tumor-associated antigens and thus promotes antigen processing. Subsequent treatment with talabostat might then promote the maturation of professional antigen-presenting cells and the stimulation of antigen-specific T cells, including cytolytic T lymphocytes, via the upregulation of cytokine and chemokine production within the tumor and lymphoid tissues.

3. Preclinical studies

In preclinical studies, talabostat demonstrates significant antitumor effects, both as a single agent and in combination with chemotherapy or monoclonal antibodies [6-8] .

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.

Page 2: Talabostat

Talabostat

1460 Expert Opin. Investig. Drugs (2007) 16(9)

Treatment of mice with talabostat inhibited the growth of a variety of tumors, including WEHI 164 (fibrosarcoma), EL4 (lymphoma) and B16 (melanoma). Long-lived and specific immunity to secondary rechallenge was also established in talabostat-treated mice that rejected their tumors. Examination of mRNA levels indicated that talabostat increased expression in the tumors and draining lymph nodes of cytokines and chemokines that have the ability to promote antitumor responses involving both innate and adaptive immunity.

4. Clinical studies

Talabostat has been studied in a variety of dose-finding studies in both healthy volunteers and patients with malignant disease.

4.1 Phase I studies in healthy volunteers Two placebo-controlled Phase I studies were conducted in healthy male volunteers. Talabostat was studied at single daily doses from 10 to 2400 µ g and at multiple single daily doses from 25 to 1800 µ g [9] . Overall, the drug was well tolerated, with sensation of temperature change being the most common complaint (25/90; 28%). In the single-dose study, tachycardia (12/54; 22%) and dizziness (8/54; 15%) were also noted; the multiple-dose study found headache (19/36; 53%), myalgia (9/36; 25%), nausea (6/36; 17%), vomiting (5/36; 14%), peripheral edema (5/36; 14%), rigors (5/36; 14%) and arthralgia (4/36; 11%).

Talabostat was rapidly absorbed, with a median time to maximum concentration (T max ) of ≤ 2 h. Maximum observed plasma concentration (C max ) values appeared to be dose proportional and increased with higher doses, as shown in Figure 3 . Once peak plasma concentrations were reached, plasma elimination of talabostat was slow. It is thought that talabostat binds irreversibly to plasma proteins and that this binding is responsible for the long half-life. Urinary excretion and renal clearance increase with dose. Plasma levels of talabostat in the multiple-dose study suggested a multi-compartmental distribution.

In the single-dose study, plasma DPP-IV activity was inhibited, in a dose-related fashion ( Figure 4 ), to ∼ 20% of baseline within 1 h and remained ∼ 30% of baseline throughout 24 h post-dose. In the multiple-dose study, doses of ≥ 100 µ g of talabostat produced pronounced inhibition of plasma DPP-IV activity to ≤ 20% of baseline

BOH

OH

N

O

NH2.CH3SO3H

Figure 1. Chemical structure of talabostat.

within 1 h post-dose, and at doses of ≥ 300 µ g, CD26/DPP-IV activity was almost completely inhibited ( > 95%) within 30 min of the first dose on day 1.

In the single-dose study, higher doses of talabostat (1200, 1800 or 2400 µ g) increased plasma IL-6 levels ∼ 10-fold at 8 h after dosing, whereas IL-11 and granulocyte colony stimulating factor (G-CSF) concentrations did not change.

In the multiple-dose study, plasma concentrations of both G-CSF and IL-6 increased as talabostat dosing was increased, with a dose- and time-dependent increase in plasma concentrations of IL-6 between day 1 and 6, with maximum plasma concentration achieved in the 1200- µ g dose level at ∼ 20 pg/ml at 6 h post-dose on day 7.

In additional Phase I trials, food had only a slight effect on the rate of absorption of talabostat, as evidenced by a 20 – 30% reduction in prolonged T max and mean C max in the presence of food. However, food had no effect on the extent of talabostat absorption, as reflected in relative bioavailability, which was in the range of 1.1 – 1.2. Similarly, whereas ingestion of antacids with talabostat decreased mean C max by apparently 30% at lower doses of drug, relative bioavailability remained in the range of 1.0 – 1.1. Following administration of talabostat 400 µg, antacid decreased the extent of absorption slightly as both C max ratio and relative bioavailability were reduced to 0.7.

In summary, these studies demonstrate that talabostat is readily absorbed, even in the presence of food or antacids, and displays predictable pharmacokinetics. It should be noted that a classic maximum tolerated dose was not defined in any of these studies.

4.2 Phase I studies in patients Talabostat was also investigated in several dose-finding studies in patients with malignant melanoma.

4.2.1 Effect of talabostat on neutrophil production in patients receiving myelosuppressive chemotherapy The ability of talabostat to induce G-CSF and thereby increase neutrophil production was evaluated in a Phase I dose-finding study of talabostat in patients receiving myelosuppressive chemotherapy [10] . Total daily talabostat doses of 200, 400, 800 and 1200 µ g (divided twice daily) were administered to patients for 7 days following administration of their second cycle of chemotherapy. Cycle 1 (chemotherapy alone) served as the patient ’ s own control. Of 13 patients receiving talabostat 800 µ g, 5 experienced a ≥ 2-day improvement in ≥ grade 3 neutropenia. A corresponding upregulation in G-CSF, IL-6 and IL-8 was observed in most patients. Overall, talabostat was generally well tolerated and a maximum tolerated dose was not reached. Adverse events reported con-siderably more frequently in patients receiving talabostat in cycle 2 compared with cycle 1 (control) were: edema/peripheral swelling (15/34 [44%] versus 1/34 [3%]); hypotension (including orthostatic) (8/34 [23%] versus 4/34 [12%]); syncope (4/34 [12%] versus 1/34 [3%]); and hypovolemia

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.

Page 3: Talabostat

Cunningham

Expert Opin. Investig. Drugs (2007) 16(9) 1461

Targeted FAP inhibition Targeted inhibition of DPP VIII and IXleads to immunostimulation

Tumor stromal fibroblasts( FAP expression)

Macrophage( DPP VIII/IX)

FAP-mediatedintravasation intocirculation

Talabostat

FAP breakdownof extracellularmatrix

FAP activationof latentgrowth factors

Induction ofFAP by tumor

Stromalfibroblasts

Innate immunity Acquired immunity

Neutrophils

Chemokines Chemokines

Tumor

Macrophages Natural killercells

CytolyticT lymphocytes

Memorycells

Cytokines andchemokines

Inhibition of DPP VIII/IXby talabostat induces

an IL-1β response(via caspase-1) in thestroma of tumor and

lymph nodes

Tumor

Talabostat

IL-1β

Figure 2. The novel dual mechanism of action of talabostat.DPP: Dipeptidyl peptidase; FAP: Fibroblast activation protein.

1200600300100250

5

10

15

20

25

30

Talabostat dose (µg)

Cm

ax (

ng

/ml)

Day 1

Day 7

Figure 3. Maximal dose tolerance study in healthy talabostat plasma levels on subjects: days 1 and 7.There were six subjects per group. For talabostat 600 µg, n = 5 as 1 subject was not dosed on day 7. Cmax: Maximal plasma concentration.

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.

Page 4: Talabostat

Talabostat

1462 Expert Opin. Investig. Drugs (2007) 16(9)

(5/34 [15%] versus 0). Two patients experienced a grade 3 adverse event that was considered to be related to talabostat, orthostatic hypotension (800 µ g/day) and syncope (1200 µ g/day).

4.2.2 Combination study of talabostat and rituximab in indolent non-Hodgkin ’ s lymphoma A Phase I study to evaluate the safety and activity of adding talabostat to a regimen of rituximab in patients with B-cell malignancies who had previously not responded to rituximab was conducted in 20 patients [11] . Rituximab 375 mg/m 2 was administered weekly × 4. Total daily doses of talabostat 400, 600 or 800 µ g b.i.d. were administered for 6 days following each dose of rituximab. In contrast to the results of the healthy volunteer studies, the adverse event most frequently reported (12/20 patients [60%]) was edema, although fatigue (8/20 patients [40%]), dizziness (7/20 patients [35%]) and nausea (7/20 patients [35%]) were also reported frequently. All adverse events were manageable and reversible following interruption or discontinuation of talabostat and/or rituximab. A maximum-tolerated dose was not determined per protocol; however, 300 µ g b.i.d. was considered the best-tolerated dose for additional study in combination with rituximab. Significant indications of clinical activity were also seen with three patients demon-strating a partial response. Elevations in the following cytokines were reported across all doses following talabostat administration: G-CSF (13/15), IL-1 β (10/15), IL-2 (7/15), IL-6 (8/15), IL-8 (8/15), IL-10 (11/15), TNF- α (11/15) and IFN- γ (3/15).

4.3 Phase II studies All of the data discussed in this section are preliminary results, as data from these studies are being verified at present.

4.3.1 Metastatic melanoma Two studies in stage IV melanoma have completed enrollment, and patients are being followed for survival. The first is a single-agent study [12] ; the second is a combination study [13] .

4.3.1.1 Study of talabostat as a single agent This study was an open-label, single-arm, two-stage multicenter trial conducted to evaluate the activity of talabostat alone in patients with stage IV melanoma with no more than one prior bio- or chemotherapy regimen. Talabostat 300 µ g b.i.d. p.o. was administered on days 1 – 14 of each 3-week cycle with dose escalation to 400 µ g b.i.d. allowed in cycle 2 or subsequent cycles, depending on tolerability. A total of 42 patients were enrolled (27 men and 15 women; median age: 51 years). The majority of patients were M1c (visceral metastases) and had received prior biotherapy or chemotherapy for stage IV disease. Of these, 31 patients were evaluable for response (evaluability defined as completion of at least two cycles of treatment with a postbaseline disease assessment). Of 31 evaluable patients, 2 demonstrated a response to treatment (per the Response Evaluation Criteria in Solid Tumors [RECIST]), including 1 patient with a durable complete response. Both responding patients were classified as M1a (skin metastases) and had

0

25

50

75

100

125

0 1 2 3 4 5 6 7 8 9 10 11 12Time post-dose on day 1 (h)

Bas

elin

e D

PP

-IV

act

ivit

y (%

)

Placebo

25 µg

100 µg

300 µg

600 µg

1200 µg

Figure 4. Plasma DPP-IV activity on study day 1 (study CA 168-002; % baseline).Note:1800 µg dose cohort not analyzed.DPP: Dipeptidyl peptidase.

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.

Page 5: Talabostat

Cunningham

Expert Opin. Investig. Drugs (2007) 16(9) 1463

progressive disease following prior cytokine treatment. The Kaplan–Meier (K–M) estimate for median progression-free survival (PFS) in all patients was 1.5 months. Median overall survival was estimated at 7.1 months. Again, the most frequently reported adverse events were edema, fatigue, hypotension and nausea, with the most com-mon grade 3 adverse event being peripheral edema. Two grade 4 events were reported: acute (pre) renal failure and aspiration pneumonia.

4.3.1.2 Combination study of talabostat/cisplatin This second study was conducted to assess the antitumor activity of the combination of talabostat and cisplatin in patients with stage IV melanoma [14] . The treatment regimen involved 21-day cycles with administration of cisplatin (75 mg/m 2 , i.v., on day 1 of cycles 1 – 4) and talabostat 300- µ g tablets b.i.d. p.o. on days 2 – 15. Dose escalation to talabostat 400 µ g b.i.d. was allowed depending on tolerability. After cycle 4, single-agent talabostat could be continued if the investigator deemed this to be appropriate. Of the 74 patients who entered the study (50 men, 24 women; median age: 58 years), most were classified as M1c. The most frequent adverse events were nausea, fatigue and vomiting and the most frequent grade 3 toxicities were vomiting, thrombocytopenia, fatigue, edema and neutropenia (4.1%). Grade 4 thrombo-cytopenia was reported in two patients and grade 4 hypotension in one patient. A total of 43 patients were evaluable for response. A partial response was reported in six patients. The median K-M estimate of PFS in all patients was 2.8 months. The median K-M estimate for overall survival in the intent-to-treat population was 8.5 months.

4.3.2 Non-small-cell lung cancer This open-label, single-arm, multicenter combination Phase II trial was conducted to evaluate the activity of talabostat and docetaxel in patients with stage IIIB/IV NSCLC who failed a platinum-based therapy [15] . The treatment regimen included docetaxel 75 mg/m 2 (day 1 of each 21-day cycle) with appropriate premedication; talabostat 200 µ g b.i.d. p.o. days 2 – 15; with dose escalation of talabostat to 300 µ g b.i.d. allowed in subsequent cycles depending on tolerability. Single-agent talabostat continued past six cycles at the discretion of the investigator. A total of 55 patients (26 men and 29 women; median age 61 years) were enrolled in the study. Of these, 35 patients had received one prior regimen for advanced NSCLC and 20 received at least two prior regimens. Of the 55 patients enrolled, 42 met evaluability criteria for response; 6 patients had a response per the World Health Organization, including 2 complete responses. K-M estimates for all patients were 4.2 months for median PFS and 8.4 months for survival. The most frequently reported regimen-related adverse events were again edema, fatigue and neutropenia.

4.3.3 Chronic lymphocytic leukemia This combination Phase II study using talabostat and rituximab was conducted to evaluate the efficacy (response rate) of talabostat and rituximab as salvage therapy in patients with advanced chronic lymphocytic leukemia who failed fludarabine and/or rituximab [16] . The study design involved a single-arm, open-label, multicenter study in up to 54 evaluable patients. The treatment regimen (28-day treatment course) included: rituximab 375 mg/m 2 i.v. weekly for 4 weeks (study days 1, 8, 15 and 22); talabostat 300- µ g tablets b.i.d. on study days 2 – 7, 9 – 14, 16 – 21 and 23 – 28; and additional courses permitted depending on tolerability and response. So far, 48 patients have entered this study. The majority of patients are men and the median age is 64.9 years. Most patients were Rai stage III or IV. The majority of patients have been treated previously with a rituximab regimen, and many have also received alemtuzumab. Of the 48 patients enrolled, 42 meet evaluability criteria. Clinical responses have been reported in 8 out of 42 evaluable patients. A total of six of eight responding patients had received prior rituximab and three had also received alemtuzumab. The most frequently reported adverse events (all toxicity grades) are peripheral edema, nausea, fever, dyspnea and fatigue.

4.4 Phase III studies Based on the Phase I and II studies discussed above, two randomized Phase III studies in NSCLC are being conducted at present. These are talabostat with docetaxel and talabostat with pemeterxed.

5. Conclusions

Talabostat is well tolerated, with peripheral edema the most notable toxicity clearly attributable to the drug. The etiology of this edema is unknown, but may be related to an increase in cytokines such as IL-6. Preliminary data from Phase II studies demonstrate clear and promising activity, particularly in NSCLC. This has prompted further evaluation in ongoing Phase III trials.

6. Expert opinion

Tumors possess a remarkable ability to organize their neighboring fibroblasts and endothelial cells into a nuturing domain [17] and so research has started to explore how this process might be disrupted. One target is FAP, a type II membrane protein found on reactive stromal fibroblasts present in epithelial cancers [18] and in granulation tissue during wound healing [3,19,20] , but is generally absent from most normal adult human tissues. The protein is conserved throughout chordate evolution, with homologs in mouse [21,22] and Xenopus laevis [23] , whose expression correlates with tissue remodeling events [20] . Why tumor-associated fibroblasts express FAP is not known, but it is interesting that FAP is induced by TGF- β [24] , which is commonly produced

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.

Page 6: Talabostat

Talabostat

1464 Expert Opin. Investig. Drugs (2007) 16(9)

by many tumors [25] . Although not involved in the development of malignancy [26] , the protein has gelatinase activity [20] and so was originally proposed to play a role in the ability of malignant cells to disrupt the extracellular matrix, and its inhibition has therapeutic effects [27,28] . In this respect, FAP does promote tumor growth in animal models [27] and so has been a candidate for therapeutic efforts with antibodies [29,30] . Thus talabostat represents the first small molecule that interacts with this intriguing protein to be actually administered to patients.

However, surprisingly, talabostat also enhances the production and release of a number of cytokines and chemokines, including IL-6, G-CSF, TNF- α , IL-10 and IL-8 from bone marrow stromal cells [6] . The proteolytic activity of peptidases such as DPP-IV/CD26 is involved in regulating the activities of polypeptide hormones and chemokines [31] , so their inhibition may alter cytokine levels and increases the chances that immune recognition of tumor associated antigens will occur. The concept of antigen-stimulatory signal proximity has been exploited in clinical studies using autologous and allogeneic tumor cells genetically modified to increase immune recognition [32-37] . However,

talabostat seems to achieve the same effect in vivo and while chemotherapy is being administered.

Perhaps most remarkably, talabostat achieves these biologic effects with an orally administered compound. This gives optimism that greater understanding of the structure and function of biologically important proteins will increasingly lead to better design of small-molecule modifiers that can be easily manufactured and administered.

Acknowledgements

The author wishes to thank JF Tomera, Director of Clinical Development at Point Therapeutics of Boston, Massachusetts for assimilating details contained within the manuscript.

Declaration of interest

C Cunningham has served on the advisory board for Point Therapeutics. This article was independently commissioned and no fee has been received for preparation of the manuscript.

Bibliography 1. BACHOVCHIN WW, PLAUT AG,

FLENTKE GR, LYNCH M, KETTNER CA: Inhibition of IGA1 proteinases from Neisseria gonorrhoeae and Hemophilus infl uenzae by peptide prolyl boronic acids. J. Biol. Chem. ( 1990 ) 265 (7): 3738 -3743.

2. COUTTS SJ, KELLY TA, SNOW RJ et al. : Structure–activity relationships of boronic acid inhibitors of dipeptidyl peptidase IV. 1. Variation of the P2 position of XAA-boropro dipeptides. J. Med. Chem. ( 1996 ) 39 (10): 2087 -2094.

3. SCANLAN MJ, RAJ BK, CALVO B et al. : Molecular cloning of fi broblast activation protein α , a member of the serine protease family selectively expressed in stromal fi broblasts of epithelial cancers. Proc. Natl. Acad. Sci. USA ( 1994 ) 91 (12): 5657 -5661.

4. LYNN KR: The isolation and some properties of dipeptidyl peptidases II and IIIfrom porcine spleen. Int. J. Biochem. ( 1991 ) 23 (1): 47 -50.

5. UNDERWOOD R, CHIRAVURI M, LEE H et al. : Sequence, purifi cation, and cloning of an intracellular serine protease, quiescent cell proline dipeptidase. J. Biol. Chem. ( 1999 ) 274 (48): 34053 -34058.

6. JONES B, ADAMS S: Characterization of innate effector cells involved in the anti-tumor activity of talabostat (PT-100).

AACR Meeting Abstr. ( 2006 ) 2006 (1): 1094 -B.

7. ADAMS S, MILLER G, JONES B: T-cell independent anti-tumor activity of talabostat (PT-100) as a single agent and in combination with chemotherapy in mice. AACR Meeting Abstr. ( 2005 ) 2005 (1): 153 -A.

8. ADAMS S, MILLER GT, JESSON MI et al. : PT-100, a small molecule dipeptidyl peptidase inhibitor, has potent antitumor effects and augments antibody-mediated cytotoxicity via a novel immune mechanism. Cancer Res. ( 2004 ) 64 (15): 5471 -5480.

9. UPRICHARD MJ, JONES B: Phase I rising multiple-dose study of talabostat (PT-100) in healthy subjects. ASH Ann. Meeting Abstr. ( 2004 ) 104 (11): 4215 .

10. NEMUNAITIS J, VUKELJA SJ, RICHARDS D et al. : Phase I trial of PT-100 (PT-100), a cytokine-inducing small molecule, following chemotherapy for solid tumor malignancy. Cancer Investig. ( 2006 ) 24 (6): 553 -561.

11. AL-KATIB A, HURD DD, RAJU R et al. : Phase I study of talabostat and rituximab in patients with indolent non-Hodgkin ’ s lymphoma with primary resistance to or progression following rituximab. ASH Annu. Meeting Abstr. ( 2004 ) 104 (11): 1403 .

12. REDMAN BG, ERNSTOFF MS, GAJEWSKI TF et al. : Phase II trial of talabostat in stage IV melanoma. ASCO Meeting Abstr. ( 2005 ) 23 (16 Suppl.): 7570 .

13. UPRICHARD MJ, O DAY SJ, PAVLICK AC et al. : Phase II study of talabostat and cisplatin in stage IV melanoma. ASCO Meeting Abstr. ( 2005 ) 23 (16 Suppl.): 7563 .

14. CUNNINGHAM C, PAVLICK AC, KHAN KD et al. : Phase II trial of talabostat and cisplatin in patients with stage IV melanoma. J. Clin. Oncol. (Meeting Abstracts) ( 2006 ) 24 (18 Suppl.): 8040 .

15. CUNNINGHAM C, RICHARDS D, SALGIA R et al. : Phase II trial of talabostat and docetaxel in patients with stage IIIB/IV NSCLC. ASCO Meeting Abstr. ( 2005 ) 23 (16 Suppl.): 7120 .

16. KHAN KD, O ’ BRIEN S, RAI KR et al. : Phase II study of talabostat and rituximab in fl udarabine/rituximab-resistant or refractory patients with CLL. J. Clin. Oncol. (Meeting Abstracts) ( 2006 ) 24 (18 Suppl.): 6598 .

17. KOUKOURAKIS MI, GIATROMANOLAKI A, HARRIS AL, SIVRIDIS E: Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.

Page 7: Talabostat

Cunningham

Expert Opin. Investig. Drugs (2007) 16(9) 1465

survival role for tumor-associated stroma. Cancer Res. ( 2006 ) 66 (2): 632 -637.

18. RETTIG WJ, GARIN-CHESA P, HEALEY JH et al. : Regulation and heteromeric structure of the fi broblast activation protein in normal and transformed cells of mesenchymal and neuroectodermal origin. Cancer Res. ( 1993 ) 53 (14): 3327 -3335.

19. GARIN-CHESA P, OLD LJ, RETTIG WJ: Cell surface glycoprotein of reactive stromal fi broblasts as a potential antibody target in human epithelial cancers. Proc. Natl. Acad. Sci. USA ( 1990 ) 87 (18): 7235 -7239.

20. PARK JE, LENTER MC, ZIMMERMANN RN et al. : Fibroblast activation protein, a dual specifi city serine protease expressed in reactive human tumor stromal fi broblasts. J. Biol. Chem. ( 1999 ) 274 (51): 36505 -36512.

21. NIEDERMEYER J, ENENKEL B, PARK JE et al. : Mouse fi broblast-activationprotein-conserved FAP gene organization and biochemical function as a serine protease. Eur. J. Biochem. ( 1998 ) 254 (3): 650 -654.

22. NIEDERMEYER J, GARIN-CHESA P, KRIZ M et al. : Expression of the fi broblast activation protein during mouse embryo development. Int. J. Dev. Biol. ( 2001 ) 45 (2): 445 -447.

23. BROWN DD, WANG Z, FURLOW JD et al. : The thyroid hormone-induced tail resorption program during xenopus laevis metamorphosis. Proc. Natl. Acad. Sci. USA ( 1996 ) 93 (5): 1924 -1929.

24. RETTIG WJ, SU SL, FORTUNATO SR et al. : Fibroblast activation protein: purifi cation, epitope mapping and induction by growth factors. Int. J. Cancer ( 1994 ) 58 (3): 385 -392.

25. KONG FM, ANSCHER MS, MURASE T et al. : Elevated plasma transforming growth factor- β 1 levels in breast cancer patients decrease after surgical removal of the tumor. Ann. Surg. ( 1995 ) 222 (2): 155 -162.

26. NIEDERMEYER J, KRIZ M, HILBERG F et al. : Targeted disruption of mouse fi broblast activation protein. Mol. Cell Biol. ( 2000 ) 20 (3): 1089 -1094.

27. CHENG JD, DUNBRACK RL Jr, VALIANOU M et al. : Promotion of tumor growth by murine fi broblast activation protein, a serine protease, in an animal model. Cancer Res. ( 2002 ) 62 (16): 4767 -4772.

28. CHENG JD, VALIANOU M, CANUTESCU AA et al. : Abrogation of fi broblast activation protein enzymatic activity attenuates tumor growth. Mol. Cancer Ther. ( 2005 ) 4 (3): 351 -360.

29. MERSMANN M, SCHMIDT A, RIPPMANN JF et al. : Human antibody derivatives against the fi broblast activation protein for tumor stroma targeting of carcinomas. Int. J. Cancer ( 2001 ) 92 (2): 240 -248.

30. SCHMIDT A, MULLER D, MERSMANN M et al. : Generation of human high-affi nity antibodies specifi c for the fi broblast activation protein by guided selection. Eur. J. Biochem. ( 2001 ) 268 (6): 1730 -1738.

31. VON BONIN A, HUHN J, FLEISCHER B: Dipeptidyl-peptidase IV/CD26 on T cells: analysis of an alternative T-cell activation pathway. Immunol. Rev. ( 1998 ) 161 : 43 -53.

32. NEMUNAITIS J, STERMAN D, JABLONS D et al. : Granulocyte-macrophagecolony-stimulating factor gene-modifi ed autologous tumor vaccines in

non-small-cell lung cancer. J. Natl. Cancer Inst. ( 2004 ) 96 (4): 326 -331.

33. O ’ ROURKE MG, SCHMIDT CW,O’ROURKE TR, ELLEM KA: Immunotherapy, including gene therapy, for metastatic melanoma. Aust. NZ J. Surg. ( 1997 ) 67 (12): 834 -841.

34. BARRIO MM, DE MOTTA PT, KAPLAN J et al. : A Phase I study of an allogeneic cell vaccine (vaccimel) with GM-CSF in melanoma patients. J. Immunother. ( 2006 ) 29 (4): 444 -454.

35. HABAL N, GUPTA RK, BILCHIK AJ et al. : Cancervax, an allogeneic tumor cell vaccine, induces specifi c humoral and cellular immune responses in advanced colon cancer. Ann. Surg. Oncol. ( 2001 ) 8 (5): 389 -401.

36. NEMUNAITIS J, JAHAN T, ROSS H et al. : Phase I/II trial of autologous tumor mixed with an allogeneic gvax vaccine in advanced-stage non-small-cell lung cancer. Cancer Gene Ther. ( 2006 ) 13 (6): 555 -562.

37. SIMONS JW, SACKS N: Granulocyte-macrophage colony-stimulating factor-transduced allogeneic cancer cellular immunotherapy: the gvax(R) vaccine for prostate cancer. Urol. Oncol. ( 2006 ) 24 (5): 419 -424.

Affi liation Charles Casey Cunningham MD Mary Crowley Medical Research Center, 1717 Main Street, 60th fl oor, Dallas, Texas, 75201, USATel: + 1 903 839 3999 ; Fax: + 1 903 839 3999 ; E-mail: [email protected]

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.

Page 8: Talabostat

Exp

ert O

pin.

Inv

estig

. Dru

gs D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

12/0

4/13

For

pers

onal

use

onl

y.


Recommended