Associate editor: P. Dent
PIM kinase (and Akt) biology and signaling in tumors

https://doi.org/10.1016/j.pharmthera.2015.03.001Get rights and content

Abstract

The initiation and progression of human cancer is frequently linked to the uncontrolled activation of survival kinases. Two such pro-survival kinases that are commonly amplified in cancer are PIM and Akt. These oncogenic proteins are serine/threonine kinases that regulate tumorigenesis by phosphorylating substrates that control the cell cycle, cellular metabolism, proliferation, and survival. Growing evidence suggests that cross-talk exists between the PIM and Akt kinases, indicating that they control partially overlapping survival signaling pathways that are critical to the initiation, progression, and metastatic spread of many types of cancer. The PI3K/Akt signaling pathway is activated in many human tumors, and it is well established as a promising anticancer target. Likewise, based on the role of PIM kinases in normal and tumor tissues, it is clear that this family of kinases represents an interesting target for anticancer therapy. Pharmacological inhibition of PIM has the potential to significantly influence the efficacy of standard and targeted therapies. This review focuses on the regulation of PIM kinases, their role in tumorigenesis, and the biological impact of their interaction with the Akt signaling pathway on the efficacy of cancer therapy.

Introduction

Solid and hematopoietic cancers utilize intercellular signaling cascades mediated by oncogenic kinases to maintain tumor cell growth and survival. In normal cells, the activity of these kinases is tightly controlled, whereas their sustained activation promotes apoptotic resistance and uncontrolled proliferation. PIM and Akt are serine/threonine kinases that are frequently activated in human cancers, and they phosphorylate overlapping substrates to activate common pathways that control various physiological processes that ultimately dictate the balance between cell survival and apoptosis. As a result, these kinases represent promising targets for cancer therapy and they are the focus of intense drug development efforts. Understanding how these kinases control distinct and overlapping signal transduction pathways is important for designing rational drug combinations and improving the efficacy of PIM and Akt inhibitors in the clinic.

The Proviral Integration site for Moloney murine leukemia virus (PIM) kinases were initially discovered through a viral-insertion screen for genes that enhance the development of lymphoma in the Eu-myc tumor model. The PIM family of serine/threonine kinases consists of three isoforms (PIM1, PIM2, and PIM3) that are highly conserved throughout evolution (Nawijn et al., 2011). Separate genes located on different human chromosomes encode the PIM isoforms, and they share high sequence homology at the amino acid level; PIM1 and PIM2 are 61% identical and PIM1 and PIM3 are 71% identical (Baytel et al., 1998). Multiple reports have demonstrated functional redundancy between PIM kinase isoforms both in vitro and in vivo (Mikkers et al., 2004, Bullock et al., 2005, Narlik-Grassow et al., 2012). Alternative translation initiation sites have been described for PIM1 and PIM2, giving rise to isoforms with different molecular weights. The PIM1 kinase encodes 33 and 44 kDa isoforms that display comparable kinase activity (Saris et al., 1991). However, the longer isoform of PIM1 was demonstrated to bind to the SH3 domain of the ETK/BMX tyrosine kinase, localizing it to the plasma membrane, whereas the shorter isoform is primarily localized in the cytosol and nucleus (Xie et al., 2006). PIM2 exists as three isoforms (34, 37, and 40 kDa), but no differences in protein function or interaction have been described. PIM3 has only been described as a single isoform (Nawijn et al., 2011). The PIM kinase isoforms are ubiquitously expressed, although some tissue specificity has been described: PIM1 is highly expressed in hematopoietic cells, gastric, head and neck, and prostate tumors (Bachmann et al., 2006, Cibull et al., 2006); PIM2 is highly expressed in lymphoid and brain tissues (Cohen et al., 2004, Mikkers et al., 2004); and PIM3 is highly expressed in breast, kidney and brain tissues (Feldman et al., 1998). In contrast to a majority of serine/threonine kinases, including Akt, the activity of PIM kinases is not regulated by cellular localization or post-translational modification. X-ray crystallography structural analyses of PIM1 revealed that this protein contains a conserved catalytic domain but lacks a regulatory domain (Qian et al., 2005). Therefore, it is thought that PIM kinases are constitutively active when expressed in cells, and their activity is directly correlated with their expression level.

The PKB/Akt family of serine/threonine protein kinases play a central role in signaling downstream of phosphatidylinositol 3-kinase (PI3K) (Cantley, 2002). The Akt gene was originally discovered as a transforming retrovirus isolated from the AKR mouse strain, which spontaneously developed many types of cancer. Subsequently, the oncogene encoding this virus was discovered and termed v-Akt. Thus, the later identified human analogues were named accordingly (Staal, 1987). Akt is a member of the AGC kinase family, which mediate a wide array of important cellular functions, and whose dysregulation is strongly associated with the pathogenesis of many human diseases, most notably cancer (Pearce et al., 2010). The activation of Akt is controlled by phosphorylation at two conserved residues known as the activation loop, which is controlled by the upstream kinase PDK-1 (phosphoinositide dependent kinase-1) (Toker & Newton, 2000), and the hydrophobic motif, which is regulated by the mTORC2 complex (Sarbassov et al., 2005a, Sarbassov et al., 2005b), as well as other kinases (Warfel et al., 2011, Xie et al., 2011); phosphorylation at both of these sites is necessary maximal activation of the enzyme. Once activated, Akt phosphorylates defined substrates throughout the cell, ultimately inducing pro-proliferation and anti-apoptotic signaling pathways (Cantley, 2002). The structure and function of Akt have been reviewed in great detail (see Vivanco and Sawyers, 2002, Hanada et al., 2004).

Section snippets

Transcriptional regulation

The expression of PIM kinases is largely regulated at the transcriptional level. The PIM kinases are downstream of multiple oncogenic tyrosine kinase receptors, including Janus kinase (JAK) (Wernig et al., 2008) and FMS-like tyrosine kinase 3 (FLT3) (Kim et al., 2005). The JAK/STAT pathway plays a critical role in regulating the expression of PIM genes. The JAK/STAT pathway represents an alternative to the second messenger signaling system, which is required for the activation of Akt, PKC, and

Proviral integration site for Moloney murine leukemia virus and protein kinase B kinase signaling in cancer

An investigation of the amino acid sequence specificity of PIM1 substrates revealed that PIM1 prefers to phosphorylate peptides with the following motif: K/R–X–X–X–S/T–X (X is neither a basic group nor a large hydrophobic residue) (Friedmann et al., 1992). Further screening of peptide libraries identified the consensus sequence that bound to PIM kinases, ARKRRRHPSGPPTA (termed “pimtide”) (Bullock et al., 2005). Notably, this consensus phosphorylation motif is strikingly similar to that of Akt

Proviral Integrations of Moloney virus kinases promote carcinogenesis

Anton Berns and colleagues originally identified the pim1 gene as an integration site of the Moloney murine leukemia virus during a screen of viral carcinogenesis (Selten et al., 1984), and all three PIM isoforms were identified as genes co-activated with myc in murine lymphoid tumors (Nawijn et al., 2011). The Pim kinases are overexpressed in a wide variety of human tumors of both hematological and epithelial origin. PIM1 expression is correlated with tumor aggressiveness, and it is a marker

Summary and perspectives

In many tumor types, prosurvival kinases represent a sought after target for drug development. While the PI3K/Akt pathway inhibitors have been validated in preclinical and clinical trials, resistance remains a major obstacle to their effective use. Based on their activity in the cell cycle and apoptosis, their frequent overexpression in cancer and their association with enhanced tumor growth and chemoresistance, the PIM kinases represent a promising target for anticancer drug discovery. As PIM

Conflict of interest

The authors disclose no conflicts of interest.

References (117)

  • N. Fujita et al.

    Akt-dependent phosphorylation of p27Kip1 promotes binding to 14–3–3 and cytoplasmic localization

    J Biol Chem

    (2002)
  • A. Hahn-Windgassen et al.

    Akt activates the mammalian target of rapamycin by regulating cellular ATP level and AMPK activity

    J Biol Chem

    (2005)
  • P.S. Hammerman et al.

    Pim and Akt oncogenes are independent regulators of hematopoietic cell growth and survival

    Blood

    (2005)
  • M. Hanada et al.

    Structure, regulation and function of PKB/AKT—a major therapeutic target

    Biochim Biophys Acta

    (2004)
  • C. Hogan et al.

    Elevated levels of oncogenic protein kinase Pim-1 induce the p53 pathway in cultured cells and correlate with increased Mdm2 in mantle cell lymphoma

    J Biol Chem

    (2008)
  • M.A. Hospital et al.

    The FLT3 and Pim kinases inhibitor SGI-1776 preferentially target FLT3-ITD AML cells

    Blood

    (2012)
  • E.K. Keeton et al.

    AZD1208, a potent and selective pan-Pim kinase inhibitor, demonstrates efficacy in preclinical models of acute myeloid leukemia

    Blood

    (2014)
  • K.T. Kim et al.

    Pim-1 is up-regulated by constitutively activated FLT3 and plays a role in FLT3-mediated cell survival

    Blood

    (2005)
  • K.S. Kovacina et al.

    Identification of a proline-rich Akt substrate as a 14–3–3 binding partner

    J Biol Chem

    (2003)
  • J.A. Losman et al.

    Protein phosphatase 2A regulates the stability of Pim protein kinases

    J Biol Chem

    (2003)
  • J. Lu et al.

    Pim2 is required for maintaining multiple myeloma cell growth through modulating TSC2 phosphorylation

    Blood

    (2013)
  • B.D. Manning et al.

    Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway

    Mol Cell

    (2002)
  • S. Matikainen et al.

    Interferon-alpha activates multiple STAT proteins and upregulates proliferation-associated IL-2Ralpha, c-myc, and Pim-1 genes in human T cells

    Blood

    (1999)
  • K. Mizuno et al.

    Regulation of Pim-1 by Hsp90

    Biochem Biophys Res Commun

    (2001)
  • T. Mochizuki et al.

    Physical and functional interactions between Pim-1 kinase and Cdc25A phosphatase. Implications for the Pim-1-mediated activation of the c-Myc signaling pathway

    J Biol Chem

    (1999)
  • M. Nieborowska-Skorska et al.

    Complementary functions of the antiapoptotic protein A1 and serine/threonine kinase Pim-1 in the BCR/ABL-mediated leukemogenesis

    Blood

    (2002)
  • T. Obata et al.

    Peptide and protein library screening defines optimal substrate motifs for AKT/PKB

    J Biol Chem

    (2000)
  • K.J. Peltola et al.

    Pim-1 kinase inhibits STAT5-dependent transcription via its interactions with SOCS1 and SOCS3

    Blood

    (2004)
  • E.M. Rainio et al.

    Pim kinases are upregulated during Epstein–Barr virus infection and enhance EBNA2 activity

    Virology

    (2005)
  • R.B. Robey et al.

    Is Akt the “Warburg kinase”?—Akt–energy metabolism interactions and oncogenesis

    Semin Cancer Biol

    (2009)
  • D.D. Sarbassov et al.

    Growing roles for the mTOR pathway

    Curr Opin Cell Biol

    (2005)
  • S. Sengupta et al.

    Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress

    Mol Cell

    (2010)
  • N. Shah et al.

    Potential roles for the PIM1 kinase in human cancer — a molecular and therapeutic appraisal

    Eur J Cancer

    (2008)
  • J.H. Song et al.

    Deletion of Pim kinases elevates the cellular levels of reactive oxygen species and sensitizes to K-Ras-induced cell killing

    Oncogene

    (2014)
  • A. Toker et al.

    Cellular signaling: pivoting around PDK-1

    Cell

    (2000)
  • M. van Lohuizen et al.

    Predisposition to lymphomagenesis in Pim-1 transgenic mice: cooperation with c-myc and N-myc in murine leukemia virus-induced tumors

    Cell

    (1989)
  • Z. Wang et al.

    Phosphorylation of the cell cycle inhibitor p21Cip1/WAF1 by Pim-1 kinase

    Biochim Biophys Acta

    (2002)
  • N.A. Warfel et al.

    Disruption of the interface between the pleckstrin homology (PH) and kinase domains of Akt protein is sufficient for hydrophobic motif site phosphorylation in the absence of mTORC2

    J Biol Chem

    (2011)
  • G. Wernig et al.

    The Jak2V617F oncogene associated with myeloproliferative diseases requires a functional FERM domain for transformation and for expression of the Myc and Pim proto-oncogenes

    Blood

    (2008)
  • M. Adam et al.

    Targeting PIM kinases impairs survival of hematopoietic cells transformed by kinase inhibitor-sensitive and kinase inhibitor-resistant forms of Fms-like tyrosine kinase 3 and BCR/ABL

    Cancer Res

    (2006)
  • J.M. Adams et al.

    The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice

    Nature

    (1985)
  • J.D. Allen et al.

    Pim-2 transgene induces lymphoid tumors, exhibiting potent synergy with c-myc

    Oncogene

    (1997)
  • Z. Beharry et al.

    The Pim protein kinases regulate energy metabolism and cell growth

    Proc Natl Acad Sci U S A

    (2011)
  • L.C. Cantley

    The phosphoinositide 3-kinase pathway

    Science

    (2002)
  • M.H. Cardone et al.

    Regulation of cell death protease caspase-9 by phosphorylation

    Science

    (1998)
  • B. Cen et al.

    Elevation of receptor tyrosine kinases by small molecule AKT inhibitors in prostate cancer is mediated by Pim-1

    Cancer Res

    (2013)
  • B. Cen et al.

    The Pim-1 protein kinase is an important regulator of MET receptor tyrosine kinase levels and signaling

    Mol Cell Biol

    (2014)
  • J. Chen et al.

    Pim-1 plays a pivotal role in hypoxia-induced chemoresistance

    Oncogene

    (2009)
  • W.W. Chen et al.

    Pim family kinases enhance tumor growth of prostate cancer cells

    Mol Cancer Res

    (2005)
  • F. Cheng et al.

    KSHV reactivation from latency requires Pim-1 and Pim-3 kinases to inactivate the latency-associated nuclear antigen LANA

    PLoS Pathog

    (2009)
  • Cited by (0)

    View full text