Skip to main content
. Author manuscript; available in PMC: 2014 Dec 1.
Published in final edited form as: Trends Cell Biol. 2013 Sep 25;23(12):10.1016/j.tcb.2013.08.006. doi: 10.1016/j.tcb.2013.08.006

Table 1.

Molecular targets and pharmacologic modulators for cellular microenvironment

Signaling Pathway/
Molecular Target
Agent(s) Effect on target Cancer cell type involved
(in vivo or cell line)
INFLAMMATION
NFKB N-acetyl cysteine, Resveratrol, Sutinib Inhibitory [13] RCC, GIST [4, 5]
COX1/2 Celecoxib, Aspirin, other NSAIDs Inhibitory [6, 7] Colon polyp recurrence, Breast, Oesophageal, Gastric, Biliary [8, 9]
NOS L-NAME Inhibitory [10] Breast [10]
AP1 signaling PYC71N, JIP peptide, Resveratrol Inhibitory [2, 11, 12] Breast [13]
NRF2 Sulphoraphane, Dimethyl fumarate Activation [14] Prostate [15]
NRF2 Brusatol Inhibitory [16] Lung [16]
MMPs Marimastat, MMP3 inhibitor peptide Inhibitory [17, 18] Lung, Colon [17, 19]
TGF β signaling AP12009, LY 364947, SB 525334 Inhibitory [2022] Prostate, Glioma [23, 24]
SENESCENCE
Notch/CSL SAHM1, MK0752, RO4949079 Inhibitory [25, 26] Breast, Prostate [24, 27]
p21 Senexin A Inhibitory [28] Adenocarcinoma [28]
MEK Trametenib, PD035901, UO126, Pimasertib Inhibitory [2931] Melanoma, Lung, Colon [3234]
AUTOPHAGY
mTORC complex Rapamycin, Torin, CCI-779, RAD001, AP23573 Activation [30, 35] Glioma, Prostate, RCC [24, 36, 37]
AMPK Metformin, AICAR, A-769662 Activation [30, 38] Breast, Colon [39, 40]
BH3 ABT 737, GX15-070 Activation [41, 42] Pancreas, Leukemia [43, 44]
HDAC Sperimidine, Apicidin, SAHA, Sulphoraphane Activation [14, 45, 46] Breast, CML [4749]
Autophagosome/ly sosome Bafilomycin A1, Chloroquine Inhibitory [50, 51] Breast, Prostate, Glioma [47, 52, 53]
Vps34, PI3Kγ 3-methyl-adenine Inhibitory [50, 51] Colon, Breast [54, 55]

• This Table indicates potential molecules that might be targeted in the stroma for cancer preventive-therapeutic purposes as well as some type of cancers or cell lines where certain indicated drugs have been found to be effective, and is not meant to be a comprehensive list of inhibitors/activators.

• Note that some agents directly target the indicated molecules while others act modulating the pathway leading to the molecular target.

List of abbreviation used:

EGCG (epigallocatechin-3-gallate); Celecoxib (sulfonamide nonsteroidal anti-inflammatory drug, NSAID); NSAIDs (non-steroidal anti-inflammatory drugs); AICAR (5-amino-1-β-ribofuranosylimidazole-4-carboxamide); Vps34 (phosphatylinositol 3-kinase class III); PI3Kγ (phosphoinositide 3-kinase gamma ); Senexin A (inhibitors of p21-dependent transcription); Resveratrol (3,5,4'-trihydroxy-trans-stilbene); Sutinib (targets multiple receptor tyrosine kinases. Including all receptors for platelet-derived growth factor (PDGF-Rs) and vascular endothelial growth factor receptors); L-NAME (N-nitro-L-arginine methyl ester); Sulphoraphane (1-Isothiocyanato-4-methylsulfinylbutane, polyvalent molecule derived from vegetables, activates NRF2); SAHM1 (hydrocarbon-stapled peptides that disrupt a critical protein-protein interaction in the Notch/CSL transcription factor complex); Marimastat (BB2516, broad spectrum MMP inhibitor); Torin (mTOR kinase activity inhibitor); CCI-779, RAD001 and AP23573 (mTOR inhibitors analog to rapamycin); Brusatol (selectively inhibits NRF2 ); JIP (Jun interacting protein1); MMP3 inhibitor I (Ac-Arg-Cys-Gly-Val-Pro-Asp); Spermidine (polyamine compound, HDAC inhibitor)

COX2 (cyclooxygenase 2); TGFβ (transforming growth factor-β); mTOR (mammalian target of rapamycin); AMPK (AMP activated kinase); MEK (mitogen-activated protein kinase); MMPs (metalloproteinase); AP1 (activating complex protein 1); NFκB (nuclear factor κB); HDAC (histone deacetylase); KEAP1 (kelch-like ECH-associated protein 1); p21 (cyclin dependent inhibitor waf1); HDAC (Histone deacetylases ); BH3 (bcl2 homology domain 3); CML (chronic myeloid leukemia); RCC (renal cell carcinoma); GIST (gastrointestinal stromal tumors)

References

1

Pfeffer, U., et al. (2005) Molecular mechanisms of action of angiopreventive anti-oxidants on endothelial cells: microarray gene expression analyses. Mutat Res 591, 198–211

2

Manna, S.K., et al. (2000) Resveratrol suppresses TNF-induced activation of nuclear transcription factors NF-kappa B, activator protein-1, and apoptosis: potential role of reactive oxygen intermediates and lipid peroxidation. J Immunol 164, 6509–6519

3

Miller, S.C., et al. (2010) Identification of known drugs that act as inhibitors of NF-kappaB signaling and their mechanism of action. Biochem Pharmacol 79, 1272–1280

4

Demetri, G.D., et al. (2006) Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 368, 1329–1338

5

Motzer, R.J., et al. (2007) Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 356, 115–124

6

Karin, M. and Greten, F.R. (2005) NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol 5, 749–759

7

Mann, J.R., et al. (2005) Mechanisms of disease: Inflammatory mediators and cancer prevention. Nat Clin Pract Oncol 2, 202–210

8

Chow, L.W., et al. (2005) Current directions for COX-2 inhibition in breast cancer. Biomed Pharmacother 59 Suppl 2, S281–284

9

Algra, A.M. and Rothwell, P.M. (2012) Effects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol 13, 518–527

10

Jadeski, L.C. and Lala, P.K. (1999) Nitric oxide synthase inhibition by N(G)-nitro-L-arginine methyl ester inhibits tumor-induced angiogenesis in mammary tumors. Am J Pathol 155, 1381–1390

11

Shi, L., et al. (2012) Activation of JNK signaling mediates connective tissue growth factor expression and scar formation in corneal wound healing. PLoS One 7, e32128

12

Ngoei, K.R., et al. (2011) Characterization of a novel JNK (c-Jun N-terminal kinase) inhibitory peptide. Biochem J 434, 399–413

13

Kaoud, T.S., et al. (2011) Development of JNK2-selective peptide inhibitors that inhibit breast cancer cell migration. ACS Chem Biol 6, 658–666

14

Osburn, W.O. and Kensler, T.W. (2008) Nrf2 signaling: an adaptive response pathway for protection against environmental toxic insults. Mutat Res 659, 31–39

15

Gibbs, A., et al. (2009) Sulforaphane destabilizes the androgen receptor in prostate cancer cells by inactivating histone deacetylase 6. Proc Natl Acad Sci U S A 106, 16663––16668

16

Ren, D., et al. (2011) Brusatol enhances the efficacy of chemotherapy by inhibiting the Nrf2-mediated defense mechanism. Proc Natl Acad Sci U S A 108, 1433–1438

17

Goffin, J.R., et al. (2005) Phase I Trial of the Matrix Metalloproteinase Inhibitor Marimastat Combined with Carboplatin and Paclitaxel in Patients with Advanced Non,ÄìSmall Cell Lung Cancer. Clinical Cancer Research 11, 3417–3424

18

Fisher, J.F. and Mobashery, S. (2006) Recent advances in MMP inhibitor design. Cancer and Metastasis Reviews 25, 115–136

19

Primrose, J.N., et al. (1999) Marimastat in recurrent colorectal cancer: exploratory evaluation of biological activity by measurement of carcinoembryonic antigen. Br J Cancer 79, 509–514

20

Akhurst, R.J. and Hata, A. (2012) Targeting the TGFβ signalling pathway in disease. 1–22

21

Sawyer, J.S., et al. (2003) Synthesis and activity of new aryl- and heteroaryl-substituted pyrazole inhibitors of the transforming growth factor-beta type I receptor kinase domain. J Med Chem 46, 3953–3956

22

Grygielko, E.T., et al. (2005) Inhibition of gene markers of fibrosis with a novel inhibitor of transforming growth factor-beta type I receptor kinase in puromycin-induced nephritis. J Pharmacol Exp Ther 313, 943–951

23

Hau, P., et al. (2007) Inhibition of TGF-beta2 with AP 12009 in recurrent malignant gliomas: from preclinical to phase I/II studies. Oligonucleotides 17, 201–212

24

Efstathiou, E. and Logothetis, C.J. (2010) A new therapy paradigm for prostate cancer founded on clinical observations. Clin Cancer Res 16, 1100–1107

25

Moellering, R.E., et al. (2009) Direct inhibition of the NOTCH transcription factor complex. Nature 462, 182–188

26

Tolcher, A.W., et al. (2012) Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors. J Clin Oncol 30, 2348–2353

27

Kondratyev, M., et al. (2012) Gamma-secretase inhibitors target tumor-initiating cells in a mouse model of ERBB2 breast cancer. Oncogene 31, 93–103

28

Porter, D.C., et al. (2012) Cyclin-dependent kinase 8 mediates chemotherapy-induced tumor-promoting paracrine activities. Proc Natl Acad Sci U S A 109, 13799–13804

29

Wang, J.Y., et al. (2007) Recent advances of MEK inhibitors and their clinical progress. Curr Top Med Chem 7, 1364–1378

30

Bain, J., et al. (2007) The selectivity of protein kinase inhibitors: a further update. Biochem J 408, 297–315

31

Davies, S.P., et al. (2000) Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 351, 95–105

32

Flaherty, K.T., et al. (2012) Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med 367, 107–114

33

Martinelli, E., et al. (2013) Antitumor activity of pimasertib, a selective MEK 1/2 inhibitor, in combination with PI3K/mTOR inhibitors or with multi-targeted kinase inhibitors in pimasertib-resistant human lung and colorectal cancer cells. Int J Cancer, 0

34

Engelman, J.A., et al. (2008) Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med 14, 1351–1356

35

Faivre, S., et al. (2006) Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov 5, 671–688

36

Takeuchi, H., et al. (2005) Synergistic augmentation of rapamycin-induced autophagy in malignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors. Cancer Res 65, 3336–3346

37

Fishman, M.N., et al. (2013) Phase Ib study of tivozanib (AV-951) in combination with temsirolimus in patients with renal cell carcinoma. Eur J Cancer 49, 2841–2850

38

Steinberg, G.R. and Kemp, B.E. (2009) AMPK in Health and Disease. Physiol Rev 89, 1025–1078

39

Zakikhani, M., et al. (2006) Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Res 66, 10269–10273

40

Buzzai, M., et al. (2007) Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res 67, 6745–6752

41

van Delft, M.F., et al. (2006) The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell 10, 389–399

42

Nguyen, M., et al. (2007) Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. Proc Natl Acad Sci U S A 104, 19512- 19517

43

Martin, A.P., et al. (2009) BCL-2 family inhibitors enhance histone deacetylase inhibitor and sorafenib lethality via autophagy and overcome blockade of the extrinsic pathway to facilitate killing. Mol Pharmacol 76, 327–341

44

Wei, Y., et al. (2010) The combination of a histone deacetylase inhibitor with the Bcl-2 homology domain-3 mimetic GX15–070 has synergistic antileukemia activity by activating both apoptosis and autophagy. Clin Cancer Res 16, 3923–3932

45

Kim, H.J. and Bae, S.C. (2011) Histone deacetylase inhibitors: molecular mechanisms of action and clinical trials as anti-cancer drugs. Am J Transl Res 3, 166––179

46

Acharya, M.R., et al. (2005) Rational development of histone deacetylase inhibitors as anticancer agents: a review. Mol Pharmacol 68, 917–932

47

Kanematsu, S., et al. (2010) Autophagy inhibition enhances sulforaphane-induced apoptosis in human breast cancer cells. Anticancer Res 30, 3381–3390

48

Shao, Y., et al. (2004) Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci U S A 101, 18030–18035

49

Carew, J.S., et al. (2007) Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 110, 313–322

50

Klionsky, D.J., et al. (2012) Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 8, 445–544

51

Mizushima, N. and Komatsu, M. (2011) Autophagy: Renovation of Cells and Tissues. Cell 147, 728–741

52

Fan, Q.W., et al. (2010) Akt and autophagy cooperate to promote survival of drug-resistant glioma. Sci Signal 3, ra81

53

Degtyarev, M., et al. (2008) Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol 183, 101–116

54

Petiot, A., et al. (2000) Distinct classes of phosphatidylinositol 3'-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J Biol Chem 275, 992–998

55

Rahim, R. and Strobl, J.S. (2009) Hydroxychloroquine, chloroquine, and all-trans retinoic acid regulate growth, survival, and histone acetylation in breast cancer cells. Anticancer Drugs 20, 736–745