Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Leukemia & Lymphoma

ISSN: 1042-8194 (Print) 1029-2403 (Online) Journal homepage: https://www.tandfonline.com/loi/ilal20

PI3K/AKT/mTOR pathway in multiple myeloma:


from basic biology to clinical promise

Vijay Ramakrishnan & Shaji Kumar

To cite this article: Vijay Ramakrishnan & Shaji Kumar (2018) PI3K/AKT/mTOR pathway in
multiple myeloma: from basic biology to clinical promise, Leukemia & Lymphoma, 59:11,
2524-2534, DOI: 10.1080/10428194.2017.1421760

To link to this article: https://doi.org/10.1080/10428194.2017.1421760

View supplementary material

Published online: 11 Jan 2018.

Submit your article to this journal

Article views: 967

View related articles

View Crossmark data

Citing articles: 31 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ilal20
LEUKEMIA & LYMPHOMA
2018, VOL. 59, NO. 11, 2524–2534
https://doi.org/10.1080/10428194.2017.1421760

REVIEW

PI3K/AKT/mTOR pathway in multiple myeloma: from basic biology to clinical


promise
Vijay Ramakrishnan and Shaji Kumar
Division of Hematology, Mayo Clinic, Rochester, MN, USA

ABSTRACT ARTICLE HISTORY


Multiple myeloma (MM), a cancer of terminally differentiated plasma cells, is the second most Received 27 October 2017
common hematological malignancy. The disease is characterized by the accumulation of abnor- Revised 6 December 2017
mal plasma cells in the bone marrow that remains in close association with other cells in the Accepted 9 December 2017
marrow microenvironment. In addition to the genomic alterations that commonly occur in MM,
KEYWORDS
the interaction with cells in the marrow microenvironment promotes signaling events within the Multiple myeloma; PI3K;
myeloma cells that enhances survival of MM cells. The phosphoinositide 3-kinase (PI3K)/protein AKT; mTOR; apoptosis;
kinase B (AKT)/mammalian target of rapamycin (mTOR) is such a pathway that is aberrantly acti- signaling
vated in a large proportion of MM patients through numerous mechanisms and can play a role
in resistance to several existing therapies making this a central pathway in MM pathophysiology.
Here, we review the pathway, its role in MM, promising preclinical results obtained thus far and
the clinical promise that drugs targeting this pathway have in MM.

Introduction malignancy of plasma cells and accounts for over


12,000 deaths per year just in the USA (https://seer.
Targeting abnormal signaling cascades specific to can-
cancer.gov/statfacts/html/mulmy.html). MM is always
cer cells continues to be an area of intense research.
preceded by premalignant conditions called
With a better understanding of these signaling events
Monoclonal Gammopathy of Undetermined
and the availability of potent and specific small mol-
Significance (MGUS) and Smoldering Multiple Myeloma
ecule inhibitors targeting such events, there is a strong
(SMM) [1–4]. Several studies have attempted to better
potential for more such agents to be incorporated into
understand the factor(s) that contribute to disease
the treatment armamentarium for various cancers.
transformation. The results from such studies have
Several such targeted agents have already shown clin-
educated us on the complex nature of MM, which
ical value such as vemurafenib in B-Raf proto-onco-
now is well recognized to be heterogeneous with a
gene, serine/threonine kinase (BRAF) mutated
multi-step evolution pattern. First, cytogenetic abnor-
melanoma, ceritinib in anaplastic lymphoma kinase
malities involving the Immunoglobulin heavy chain
(ALK) rearranged non-small cell lung cancer and imati-
locus are observed in MGUS, SMM, and active MM
nib in Philadelphia chromosome positive chronic mye-
patients. Some of the commonly observed transloca-
logenous leukemia (CML), and acute lymphoblastic
tions are t(11;14), t(4;14), t(14;16), and t(14;20) resulting
leukemia (ALL). Here, we review the central role played
in elevated levels of cyclin D, FGFR3, and MMSET,
by the PI3K/AKT/mTOR pathway in multiple myeloma
c-MAF, and MAFB, respectively. SMM patients having
(MM) growth and survival, promising preclinical results
these translocations are at a greater risk of progression
from studies examining inhibitors of this pathway, and
to active MM than those with no translocations. SMM
the potential for such drugs as therapeutic agents in
patients with t(4:14) have the highest risk with a
the management of MM.
median progression time of 28 months [5–9]. t(4;14) is
significantly more prevalent in active MM when com-
Multiple myeloma – a cancer with multiple
pared with premalignant conditions whereas t(14;16) is
genetic abnormalities
significantly more prevalent in patients with plasma
MM is a devastating disease that affects around 30,000 cell leukemia when compared with premalignant con-
people annually in the USA. It is an incurable ditions and MM [6,7,10]. Second, mutations in RAS,

CONTACT Vijay Ramakrishnan Ramakrishnan.vijay@mayo.edu Mayo Clinic, Division of Hematology, Mayo Clinic, 200 First St SW, Rochester, MN
55905, USA
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
AKT PATHWAY IN MYELOMA 2525

amplification of Myc, and mutations in the NF-jB path- signal transducer and activator of transcription (Jak/
way are commonly observed in active MM but not in Stat) pathways and promote tumor growth [25]. The
MGUS and SMM and are thought to be driver muta- complexity of the PI3K/AKT/mTOR pathway is ampli-
tions and alterations for progression [11–15]. Third, fied by the presence of numerous feedback loops that
MM patients can acquire additional mutations in RAS, exist within this pathway and the crosstalk with
p53, Cereblon (CRBN), IRF4, XBP1 with disease progres- numerous other pathways.
sion or response to therapies [16]. Thus, MM quite
expectedly has been a difficult disease to treat and
Feedback loops within the PI3K/AKT/mTOR
manage given the heterogeneity observed in patients.
pathway
In spite of this, MM has had a dramatic increase in sur-
vival post incorporation of immunomodulatory drugs As stated above, IGF binds to IGF receptor (IGFR)
and proteasome inhibitors to the treatment regimen expressed on plasma cells, which then activates PI3K
[17]. However, patients continue to relapse and through the phosphorylation of insulin receptor sub-
become refractory to existing therapies. Hence, tar- strate (IRS) 1 and 2 at tyrosine (Tyr) residues. Activated
geted therapies based on mutational status or aber- PI3K then recruits pleckstrin homology (PH) domain
rant signaling events are areas of intense research in containing proteins 3-phospho inositide-dependent
MM. protein kinase 1 (PDK1) and AKT to the cell surface,
In addition to these genetic alterations, MM plasma where AKT gets phosphorylated at Threonine (Thr) 308
cells are very dependent on the supporting cells in the and activated by PDK1. Once AKT gets activated, it
bone marrow microenvironment for their growth and promotes cell proliferation, anti-apoptosis, and metab-
survival. The intimate association of the MM cells with olism signals leading to tumor growth and survival
the stromal and endothelial cells in the microenviron- [21,22,26,27]. A key downstream target of AKT is the
ment contributes to elevated levels of cytokines and mammalian target of rapamycin (mTOR) complex 1
cytokine stimulated pro-survival pathways such as the (mTORC1) that promotes protein synthesis, glycolysis
PI3K/AKT/mTOR pathway within the plasma cells that and inhibits autophagy (Figure 1). In addition, acti-
are vital for MM cells to grow, survive, and develop vated mTORC1 causes inhibitory phosphorylation of
resistance to existing therapies [18,19]. IRS1 and 2 leading to feedback inhibition of the PI3K/
AKT pathway [20,26]. mTOR exists as two distinct multi
protein complexes namely mTORC1 and mTORC2. The
PI3K/AKT/mTOR – a pathway commonly
catalytic member of both these complexes is mTOR
activated in MM
with as many as five and six other binding partners in
The PI3K/AKT/mTOR pathway has been extensively mTORC1 and mTORC2, respectively [28–36]. While
studied and we would like to direct the readers to mTORC1 is regulated by phospho (p) AKT (Thr 308), it
excellent reviews published on the pathway and its is now well known that mTORC2 is the kinase that
role in various malignancies [20–22]. In MM, activating phosphorylates AKT at the serine (Ser) 473 residue
mutations in PI3K and AKT have not been identified [20]. Once phosphorylated at the Ser 473 residue, AKT
[23]. Similarly, deficiencies in phosphatase and tensin modulates survival and cell cycle progression signals
homolog (PTEN) also occur in a very small proportion through forkhead box protein O1 (FoxO1) phosphoryl-
of MM patients [24]. In spite of all this, the pathway is ation and serum-and glucocorticoid-induced protein
activated and is of importance for MM cell survival kinase (SGK) activation [26]. Furthermore, it has been
and growth. MM is predominantly a cancer localized suggested that the mTOR complexes cross inhibit each
to the bone marrow where the cancerous plasma cells other complicating the pathway activation status [37].
remain in close direct and indirect communication In addition to the above-mentioned feedback loops,
with other non-tumor cells in the bone marrow micro- MM cells modulate the PI3K/AKT/mTOR pathway
environment like stromal and endothelial cells. These through other unique mechanisms. Peterson et al.
interactions lead to increased secretion of interleukin 6 identified an mTOR interacting protein DEP domain
(IL6) by the stromal cells and vascular endothelial containing TOR interacting protein (DEPTOR) that was
growth factor (VEGF) and insulin-like growth factor able to inhibit both mTORC1 and mTORC2 [33]. As
(IGF) by MM cells. These cytokines activate their expected, DEPTOR was expressed at low levels in vari-
respective receptors expressed on MM cells to up ous tumors. Surprisingly, DEPTOR was highly expressed
regulate signaling events such as the PI3K/AKT/mTOR, in MM cells. The authors found that overexpression of
mitogen-activated protein kinase kinase/extracellular DEPTOR inhibited mTORC1, which then relieved the
signal-regulated kinase (MEK/ERK) and janus kinase/ feedback inhibition of ribosomal protein S6 kinase-
2526 V. RAMAKRISHNAN AND S. KUMAR

Figure 1. Overview of the PI3K/AKT/mTOR pathway and the feedback loops that have been documented within this pathway in
MM. IGF1: insulin-like growth factor1; IGF1R: IGF1 receptor; VEGF: vascular endothelial growth factor; VEGFR: VEGF receptor;
PI3K: phosphoinositide 3-kinase; PDK: phosphoinositide-dependent kinase 1; AKT: protein kinase B; TSC1/2: tuberous sclerosis
protein 1/2; mTORC1: mammalian target of rapamycin complex 1; mTORC2: mammalian target of rapamycin complex 2; Raptor:
regulatory-associated protein of mTOR; DEPTOR: DEP domain containing TOR interacting protein; Rictor: rapamycin-insensitive com-
panion of mammalian target of rapamycin (RICTOR); p70S6K: ribosomal protein S6 kinase; 4EBP1: eukaryotic translation initiation
factor 4E-binding protein; PKC: protein kinase C; SGK: serum-and glucocorticoid-induced protein kinase.

insulin receptor substrate 1 (pS6K-IRS1) causing activa- other mechanisms such as high basal levels of endo-
tion of AKT and survival of MM cells (Figure 1). plasmic reticulum (ER) stress and glucose-regulated
Interestingly, DEPTOR overexpression was confined to protein 78 (GRP78) cell surface localization could also
myeloma cells with translocations involving cyclin D1, contribute to constitutively activated AKT signaling in
D3, c-MAF, or MAFB [33]. Fernandez-Saiz et al. investi- MM cells [39]. Finally, we think that epigenetic
gated how mTORC1 and 2 interacting proteins telo- changes could also contribute to activation of this
mere maintenance 2 (Tel2) and Tel2 interacting pathway in MM cells [40].
protein 1 (Tti1) modulate mTOR levels during starva-
tion [38]. In that study, they observed that during
Cross talk with other signaling pathways
energy crisis, Casein Kinase 2 (CK2) phosphorylates
Tel2 and Tti1 specifically within mTORC1 marking In addition to the feedback loops that exist within the
them for degradation by the F-box containing protein PI3K/AKT/mTOR pathway, numerous other pathways
Fbxo9. The final result due to these events was cross talk with this pathway. The most intimate and
mTORC1 inactivation and increase in mTORC2 medi- well-documented cross talk is between the PI3K/AKT/
ated AKT phosphorylation and survival (Figure 1). The mTOR and RAS/RAF/MEK/ERK pathways. Depending on
authors observed high levels of Fbxo9 expression in a the cell type and context, studies have shown that
fraction of MM patients and cell lines. Knockdown of these two pathways could cross inhibit or cross acti-
Fbxo9 decreased the mTORC2/AKT signaling and sur- vate one another. Yu et al. showed that inhibiting ERK
vival of these cells. Interestingly, Fbxo9 overexpression with the MEK inhibitor U0126 promoted the epidermal
was found to occur in hyperdiploid MM and not in the growth factor (EGF) stimulated association of Grb2
other subgroups [38]. Thus, DEPTOR and Fbxo9 over- associated binder-1 (Gab1) with PI3K thereby activat-
expression appear to be mutually exclusive events and ing the PI3K/AKT pathway [41]. Similarly, AKT activa-
represent two unique mechanisms through which the tion phosphorylated RAF in the amino terminal
PI3K/AKT/mTOR pathway gets activated in a large pro- regulatory domain, which then allows 14-3-3 protein
portion of MM patients. In addition, we suspect that to bind RAF and inhibit its functions [42,43]. To the
AKT PATHWAY IN MYELOMA 2527

contrary, other studies have shown that both path- eukaryotic translation initiating factor 4E-binding pro-
ways could cross activate each other through several tein (4EBP1) [20]. Finally, in a disease like MM where
mechanisms. RAS can directly interact with and acti- the cells have to deal with high basal levels of ER
vate PI3K [44,45]. In addition, RAS/RAF/MEK/ERK path- stress, blocking translation by mTORC1 inhibition could
way can feed into the PI3K/AKT/mTOR pathway at the promote MM cell survival by partially relieving the
mTOR level [46,47]. Moreover, PI3K has been shown to high basal ER stress. Nevertheless, mTORC1 inhibition
activate RAF through a Rac/PAK-dependent mechan- still has promise in MM in a combination setting with
ism [48]. Taken together, it is clear that these two agents that promote cell death and those that could
pathways are in a complex partnership, the outcomes block resistance mechanisms to mTORC1 inhibition.
of which can vary greatly based on cellular physiology, mTORC1 inhibitors have shown synergy when used in
stress and exposure to treatment. Steinbrunn et al. combination with existing agents such as dexametha-
[49] observed that RAS silencing did not modulate the sone, revlimid, and panobinostat and also other novel
PI3K/AKT/mTOR pathway in MM suggesting a lack of a agents such as the receptor tyrosine kinase inhibitor
direct link between these two pathways. However, sorafenib [58–62], heat shock protein 90 (HSP90)
results from preclinical studies suggest that pathway inhibitor 17-AAG [63], an IGF1R inhibitor NVP-AEW541
cross inhibition could be more common in MM since [64], and AKT inhibitor MK2206 [50] (Table 1).
inhibiting one of the pathways with a small molecule To circumvent some of the resistance mechanisms
inhibitor leads to cross activation of the other pathway triggered in response to rapalogs, PI3K/mTOR inhibi-
[50,51]. Clearly, more studies are needed to (1) under- tors and mTOR kinase inhibitors have been investi-
stand if these two pathways cross talk and (2) if the gated in MM and these agents hold more promise as
cross talk is mainly cross activating or inhibiting. In anti-MM agents. The PI3K/mTOR inhibitor BEZ235
addition to the interaction with the RAS/RAF/MEK/ERK induced potent apoptosis in several MM cell lines as a
pathway, the PI3K/AKT/mTOR pathway cross talks with single agent and also enhanced activity of existing
other oncogenic pathways as well. It was reported agents such as bortezomib, doxorubicin and melpha-
that AKT inhibition prevents the inhibitory phosphoryl- lan [65,66]. Furthermore, BEZ235 was found to pro-
ation of FOXO to up regulate receptor tyrosine kinases mote osteogenic differentiation in bone marrow
such as insulin-like growth factor receptor (IGF1R), stromal cells and hence could help in alleviating bone
insulin receptor (IR) and receptor tyrosine-protein kin- complications in MM [67]. However, it must be noted
ase erbB-3 (HER3) [52]. In another study, it was that mTORC1 inhibition relieves the inhibitory phos-
observed that PI3K/AKT inhibition led to activation of phorylation of IRS1 and 2 leading to PI3K activation
Jak2/Stat5 cascade leading to resistance [53]. The and AKT phosphorylation, which may lead to resist-
results from our studies have suggested cross talk ance to PI3K/mTOR inhibitors. Given the role of
between the PI3K/AKT/mTOR and Notch as well as mTORC2 in AKT Ser 473 phosphorylation and survival
with Jak2/Stat3 pathways [54,55]. of MM cells, mTOR kinase (mTORC1 and 2) inhibitors
have also been examined in preclinical studies. In the
first study using pp242, a mTOR kinase inhibitor,
Preclinical studies using inhibitors of the PI3K/
Hoang et al. [68] observed that pp242 could inhibit
AKT/mTOR pathway in MM
AKT phosphorylation at Ser 473 and induced more
Much of the initial preclinical studies were performed potent proliferation inhibition and apoptosis than
using the mTORC1 inhibitor rapamycin and other rapa- rapamycin. Moreover, they also observed synergistic
logs [56]. Results from such studies showed that these cytotoxicity when combined with bortezomib. It must
agents were able to induce a cytostatic but not a cyto- be noted that rapamycin was antagonistic when used
toxic response. Even though these results were disap- with bortezomib [68]. We think that pAKT inhibition by
pointing, it is now clear that the lack of activity could mTOR kinase inhibitors but not by mTORC1 inhibitors
be attributed to several factors. First, several feedback could be the main factor why mTOR kinase inhibitors
loops exist like those that we reviewed above that but not mTORC1 inhibitors synergized with bortezomib.
contribute towards resistance to mTORC1 inhibitors A few other mTOR kinase inhibitors have been investi-
[57]. Second, rapamycin and rapalogs do not cause gated and results from them consistently showed
complete inhibition of mTORC1 activity. For instance, pronounced apoptosis induction in MM cells [69,70].
rapalogs are able to completely abrogate the phos- In spite of such promising effects, one must remember
phorylation of ribosomal protein S6 kinase (p70S6K) that the feedback loop from mTORC1 to IGF1R still
and phospho ribosomal protein S6 (pS6). However, exists, which could get activated post mTOR kinase
they do not fully inhibit the phosphorylation of inhibitor treatment. This was noted in a study using
2528 V. RAMAKRISHNAN AND S. KUMAR

Table 1. Results from preclinical studies examining PI3K/AKT/mTOR pathway inhibitors in MM.
Drug Target Response Reference
Rapamycin and rapalogs Rapamycin: mTORC1 Cytostatic response [56]
Rapamycin plus Rapamycin: mTORC1 Synergistic cytotoxicity was observed. The combination [58]
lenalidomide Lenalidomide: retained activity in the presence of cytokines IL6, IGF-1
Immunomodulatory and bone marrow stromal cells
agent
Rapamycin plus Rapamycin: mTORC1 Synergistic apoptosis mediated by cap-dependent transla- [59]
dexamethasone Dexamethasone: tion inhibition. mTOR inhibition led to decreased transla-
corticosteroid tion of XIAP, cIAP1, HSP27, BAG3
RAD001 plus LBH589 RAD001:mTORC1 Synergistic cytotoxicity and proliferation inhibition was [60]
LBH589: Pan-Histone observed in cell lines and patient cells. Drug combin-
deacetylase (HDAC) ation caused G0/G1 arrest and inhibition of
inhibitor angiogenesis.
Rapamycin plus Rapamycin: mTORC1 Enhanced apoptosis was observed in cell lines and patient [61]
entinostat Entinostat: Class I cells. Entinostat caused up regulation of p21 and p16
HDAC inhibitor and the drug combination decreased cyclin D, Bcl-XL,
MAPK, survivin
Rapamycin plus 17- Rapamycin: mTORC1 Drug combination induced pronounced apoptosis, cell cycle [63]
N-allylamino-17-deme- inhibitor arrest, inhibited angiogenesis and osteoclast formation
thoxygeldanamycin 17-AAG: HSP90
(17-AAG) inhibitor
BEZ235 BEZ235: PI3K/mTORC1 BEZ235 induced both cell cycle arrest and cell death in [65–67]
inhibitor many myeloma cell lines and patient cells. Components
of the tumor microenvironment were unable to prevent
BEZ235 induced cell death. BEZ235 induced synergistic
cell death in combination with bortezomib, doxorubicin,
dexamethasone and melphalan. BEZ235 promoted osteo-
genic differentiation mediated by mTOR inhibition
pp242 mTORC1/2 pp242 induced potent apoptosis mediated by both rapamy- [68]
cin sensitive and insensitive target inhibition. pp242
induced synergistic apoptosis when combined with
bortezomib
INK128 mTORC1/2 INK128 induced more potent apoptosis than mTORC1 [69]
inhibitor rapamycin. Inhibition of p-4EBP1 and pAKT
were observed to be reliable biomarkers for mTORC2
inhibition.
AZD8055 mTORC1/2 AZD8055 treatment caused apoptosis in MM cells. The [70]
authors observed up regulation of IGF1R phosphorylation
after AZD8055 treatment and blocking IGF1R using an
IGF1R neutralizing antibody enhanced cell kill induced
by AZD8055
MK2206 AKT MK2206 induced potent apoptosis in some but not all cell [50]
lines. High baseline pAKT levels correlated with sensitiv-
ity to MK2206. pERK mediated activation of PI3K/AKT/
mTOR pathway downstream of AKT was observed to be
a factor contributing to resistance to MK2206. MK2206
induced synergistic cytotoxicity when used in combin-
ation with rapamycin, LY294002, MEK1/2 inhibitor and
dexamethasone
TAS-117 AKT High baseline pAKT levels correlated with sensitivity to TAS- [51]
117. TAS-117 further increased ER stress induced by pro-
teasome inhibitors leading to irrecoverable ER stress and
apoptosis
PI-103 PI-103: PI3K/mTOR Autophagy inhibition by bafilomycin A1 enhanced apop- [72]
Bafilomycin A1: tosis induced by PI-103
Autophagy inhibitor
BEZ235 BEZ235: PI3K/mTOR Chloroquine promoted BEZ235 induced apoptosis in MM [72]
Chloroquine: cells
Autophagy inhibitor

the mTOR kinase inhibitor AZD8055, where the drug membranes and inhibits AKT, was examined in preclin-
treatment led to up regulation of IGF1R and subse- ical studies. Perifosine induced cytotoxicity in MM cell
quently AKT phosphorylation at Ser 473. The authors lines and patient cells and synergized with bortezo-
showed that IGF1R blocking antibody was able to mib, dexamethasone, doxorubicin, melphalan, and
reverse the IGF1R induced pAKT and enhance cell MEK1/2 inhibitor U0126 [71]. We examined the anti-
death induced by AZD8055 [70] (Table 1). MM effects of an AKT-specific allosteric inhibitor
AKT inhibitors have also been evaluated in MM. MK2206 and observed that cell lines with high basal
Perifosine, an alkylphospholipid that binds to cell levels of pAKT were significantly more sensitive to
AKT PATHWAY IN MYELOMA 2529

MK2206 [50]. Moreover, we found that cells with no to apoptosis of tumor cells [74]. It remains to be seen if
low basal levels of pAKT still expressed similar levels of mTORC1 could similarly function as a tumor suppres-
pmTOR like those cells with high basal levels of pAKT. sor in MM in nutrient deprived conditions.
We identified that constitutively active pERK in MM
cells contributed to activation of mTOR [50]. Using
Clinical trials
MK2206 in combination with the MEK inhibitor U0126
induced synergistic cell death. In addition, MK2206 The results obtained from preclinical studies using inhib-
synergized with the PI3K inhibitor LY294002, rapamy- itors of the PI3K/AKT/mTOR pathway strongly supported
cin, and dexamethasone in inducing cell death in MM clinical evaluation of such agents in MM patients.
cells [50]. More recently, Mimura et al. [51] also mTORC1 inhibitors were the first to be examined.
observed that the allosteric AKT inhibitor TAS-117 In a phase II trial, Farag et al. examined the clinical
induced more potent cell death in cells expressing efficacy of temsirolimus (CCI-779) in relapsed refractory
high basal levels of pAKT. Thus, pAKT levels could MM [75] (Table 2). Sixteen patients were enrolled in
serve as a biomarker for identifying patients who are this study and received at least two cycles of treat-
likely to respond to AKT inhibitors. In addition, they ment. The response to temsirolimus was minimal with
found that TAS-117 enhanced ER stress induced by only one patient achieving a partial response (PR) and
bortezomib and carfilzomib to induce irreversible ER five achieving minimal responses (MR). The disease
stress and enhanced apoptosis in MM cells [51] remained stable in six patients and progressed in the
(Table 1). mTORC1 is the most well-known inhibitor of remaining four patients. The time to progression (TTP)
autophagy [20]. Targeted inhibition of mTORC1 could was observed to be 4 months. The authors performed
therefore result in autophagy induction, and cancer western blots on peripheral blood mononuclear cells
cell survival or death. In a disease like MM with abnor- (PBMCs) to check for target inhibition post treatment.
mally high levels of misfolded proteins, autophagy They observed correlation between p-p70S6K and
might be an important mechanism through which the p-4EBP1 inhibition in PBMCs and response to temsiroli-
cancer cells eliminate these proteins and maintain cel- mus with target inhibition observed only in patients
lular homeostasis. mTORC1 inhibition by either PI3K, who showed a MR or PR to the drug [75]. In another
AKT, or mTOR inhibitors could, therefore, promote cell study, everolimus (RAD001) was examined in a phase I
survival through induction of autophagy. Aronson trial and the result was very similar to the study by
et al. [72] observed autophagy induction post treat- Farag et al. [76,77] (Table 2). However, the investiga-
ment with a dual PI3K/mTOR inhibitor PI-103. By com- tors observed down regulation of pmTOR in plasma
bining PI-103 with an autophagy inhibitor bafilomycin cells of all patients irrespective of the lack or presence
A1, the authors showed enhanced apoptosis in MM of a response [77]. These two trials showed that
cells. The authors observed similar effects when they mTORC1 inhibitors are reasonably well tolerated but
used the PI3K/mTOR inhibitor BEZ235 in combination largely ineffective in MM. In subsequent trials, mTOR
with a different autophagy inhibitor chloroquine [72]. inhibitors were examined in combination with either
In another study, it was observed that the HSP90 bortezomib or lenalidomide. Yee et al. evaluated ever-
inhibitor 17-dimethylaminoethylamino-17-demethoxy- olimus in combination with lenalidomide in a phase I
geldanamycin-induced apoptosis (17-DMAG) promoted trial on relapsed MM patients [78] (Table 2). Out of the
autophagy by inhibiting mTORC1 [73]. The authors 26 patients evaluated, 23 were considered evaluable
observed enhanced apoptosis when they combined for response and one patient showed a complete
17-DMAG with the autophagy inhibitor 3-methylade- response (CR), four patients showed PR and 10
nine (3-MA) [73]. Examining AKT or mTOR inhibitors in achieved a MR for a response rate of 65%. The drug
combination with autophagy inhibitors could, there- combination was considered to be reasonably safe.
fore, demonstrate more promising results in MM The common grade 3 or 4 adverse events observed
patients. Although this combinatorial approach looks were found to be thrombocytopenia (35%) and neu-
promising in in vitro studies, the nutrient deprived tropenia (42%). The authors performed correlative
conditions that the tumors are exposed to in vivo studies using plasma cells from patients before and
could dramatically alter the responses observed. In a after treatment. The results showed down regulation
recently published study, it was shown that tumors of p-p70S6K and in addition gene expression profiling
grown in medium deprived of all amino acids except showed that the responders expressed higher levels of
glutamine inhibit autophagy by up regulating pmTOR pathway members than the non-responders
mTORC1 [74]. In this context, glutaminolysis activated [78]. Temsirolimus in combination with bortezomib
mTORC1, which inhibited autophagy and promoted was also evaluated in relapsed refractory MM in a
2530 V. RAMAKRISHNAN AND S. KUMAR

Table 2. Results from clinical trials using PI3K/AKT/mTOR pathway inhibitors in MM.
Phase of # of
Drug Drug target trial patients Outcomes Toxicities Reference
Temsirolimus mTORC1 Phase II 16 Partial response: 1 patient Fatigue, neutropenia, [75]
(CCI-779) Minimal response: 5 patient thrombocytopenia,
Stable disease: 6 patients anemia and stomatitis
Progressive disease: 4 patients
Time to progression: 4 months
Everolimus mTORC1 Phase I 17 Partial response: 1 patient Thrombocytopenia, [76,77]
Minimal response: 1 patient leukopenia, anemia
Stable disease: 8 patients
Everolimus þ mTORC1 þ Phase I 23 Complete response: 1 patient Thrombocytopenia and [78]
lenalidomide Immunomodulator Partial response: 4 patients neutropenia. The drug
Minimal response: 10 patients combination was con-
sidered to be reason-
ably safe.
Temsirolimus þ Phase I 20 Very good partial response: 1 patient Thrombocytopenia, lym- [79]
Bortezomib Partial response: 1 patient phopenia, neutro-
Minimal response: 2 patients penia, leucopenia,
Stable disease: 12 patients anemia and diarrhea
Progressive disease: 1 patient
Temsirolimus þ Borte- Phase II 43 Complete response: 2 patients Thrombocytopenia, lym- [79]
zomib Very good partial response: 4 phopenia, neutro-
patients penia, leucopenia,
Partial response: 8 patients anemia and diarrhea
Minimal response: 6 patients
Stable disease: 19 patients
Progressive disease: 1 patient
TAK-228 mTORC1/2 Phase I 31 Minimal response: 1 patient Thrombocytopenia, [80]
Stable disease: 14 patients fatigue, neutropenia,
Progressive disease: 11 patients anemia, nausea,
hyperglycemia
Perifosine þ AKT Phase I/II 73 Complete response: 3 patients Thrombocytopenia, neu- [81]
Bortezomib ± Partial response: 13 patients tropenia, anemia and
Dexamethasone Minimal response: 14 patients pneumonia
Stable disease: 30 patients
Perifosine þ Phase I 30 Near complete response: 4 patients Thrombocytopenia, [82]
Lenalidomide þ Very good partial response: 3 leucopenia, lympho-
Dexamethasone patients penia, hyperglycemia,
Partial response: 8 patients hypophosphatemia,
Minimal response: 7 patients arthralgia, fatigue,
Stable disease: 6 patients diarrhea, nausea
Progressive disease: 2 patients
Perifosine Phase III Discontinued due to non-AKT related
toxicities
Afuresitib AKT 34 Partial response: 3 patients Neutropenia, rash, ody- [83]
Minimal response: 3 patients nophagia, fatigue,
Stable disease: 14 patients thrombocytopenia,
nausea, diarrhea
Afuresitib þ AKT, MEK/ERK Not tolerated [83,84]
Trametinib

phase 1/2 trial [79] (Table 2). In this study, it was inhibitors [81,82]. Perifosine was examined in heavily
observed that one-third of the patients achieved at pretreated relapsed refractory MM patients in combin-
the very least a PR. Grade 3–4 adverse events ation with bortezomib with or without dexamethasone
observed were thrombocytopenia, lymphopenia, neu- in a phase I/II trial [81]. The overall response rate
tropenia, leucopenia, anemia, and diarrhea [79]. Given (ORR) defined as MR or better was 41% with 4% CR,
the promising preclinical studies seen using mTORC1/2 18% PR. The duration of response was over 6 months
inhibitors, Ghobrial et al. [80] investigated the effects for all patients showing a response. Patients who pro-
of TAK-228, a mTORC1/2 inhibitor in relapsed refrac- gressed on treatment or those with stable disease
tory MM in a phase I trial (Table 2). The results were given dexamethasone and the ORR of those
obtained were disappointing as the response to patients was 38% (2%CR, 6PR, and 10MR). The most
TAK-228 was similar to what was observed with single common grade 3 or toxicities included thrombocyto-
agent temsirolimus or everolimus. It remains to be penia, neutropenia, anemia, and pneumonia. In
seen if TAK-228 or other mTORC1/2 inhibitors demon- another study, perifosine was investigated in a phase I
strate better activity when used in combination with trial in combination with lenalidomide and dexametha-
other existing therapies. The best response yet with sone in relapsed refractory MM patients [82]. The
PI3K/AKT/mTOR pathway inhibitors has been with AKT results obtained were very promising with an ORR of
AKT PATHWAY IN MYELOMA 2531

73% and 50% of them with a PR or better. The median ORCID


progression-free survival was 10.8 months and median
Vijay Ramakrishnan http://orcid.org/0000-0003-3521-102X
overall survival was 30.6 months. Furthermore, the Shaji Kumar http://orcid.org/0000-0001-5392-9284
authors observed that response positively correlated
with increased levels of pAKT suggesting that pAKT
could serve as a biomarker in predicting responses References
[82]. Given the very promising results with perifosine, [1] Landgren O, Kyle RA, Pfeiffer RM, et al. Monoclonal
a large phase III trial was initiated (NCT01002248). gammopathy of undetermined significance (MGUS)
However, the trial was discontinued due to non-AKT- consistently precedes multiple myeloma: a prospect-
ive study. Blood. 2009;113:5412–5417.
related toxicities associated with perifosine treatment.
[2] Kyle RA, Therneau TM, Rajkumar SV, et al. A long-term
Despite the huge disappointment, perifosine demon- study of prognosis in monoclonal gammopathy of
strated the potential that AKT inhibition has in MM undetermined significance. N Engl J Med. 2002;346:
treatment and how we could target the PI3K/AKT/ 564–569.
mTOR pathway effectively to achieve meaningful [3] Kyle RA, Therneau TM, Rajkumar SV, et al. Prevalence
responses in MM. However, it must be noted that a of monoclonal gammopathy of undetermined signifi-
cance. N Engl J Med. 2006;354:1362–1369.
clinical trial using the AKT-specific inhibitor afuresertib
[4] Kyle RA, Remstein ED, Therneau TM, et al. Clinical
as a single agent showed minimal response and a trial course and prognosis of smoldering (asymptomatic)
using afuresertib in combination with the MEK inhibitor multiple myeloma. N Engl J Med. 2007;356:
trametinib was not well tolerated [83,84]. Even though 2582–2590.
targeting the PI3K/AKT/mTOR and MEK/ERK pathways [5] Fonseca R, Bailey RJ, Ahmann GJ, et al. Genomic
looked promising in preclinical studies, it appears that abnormalities in monoclonal gammopathy of undeter-
mined significance. Blood. 2002;100:1417–1424.
the dosing schedule could be crucial in minimizing toxic-
[6] Avet-Loiseau H, Facon T, Grosbois B, et al.
ities associated with such combinations [84]. Oncogenesis of multiple myeloma: 14q32 and 13q
Thus, despite promising pre-clinical data, inhibitors chromosomal abnormalities are not randomly distrib-
of the PI3K/AKT/mTOR pathway have so far seen lim- uted, but correlate with natural history, immuno-
ited clinical activity in MM. logical features, and clinical presentation. Blood.
2002;99:2185–2191.
[7] Avet-Loiseau H, Facon T, Daviet A, et al. 14q32 trans-
Conclusions locations and monosomy 13 observed in monoclonal
gammopathy of undetermined significance delineate
Basic and translational studies performed on the PI3K/ a multistep process for the oncogenesis of multiple
AKT/mTOR pathway in MM have shown that it plays myeloma. Intergroupe Francophone du Myelome.
an integral role in MM disease biology. However, it still Cancer Res. 1999;59:4546–4550.
[8] Dhodapkar MV, Sexton R, Waheed S, et al. Clinical,
remains unclear if one or multiple targets need to be
genomic, and imaging predictors of myeloma pro-
inhibited within this pathway to achieve durable gression from asymptomatic monoclonal gammopa-
responses. The various feedback loops that exist within thies (SWOG S0120). Blood. 2014;123:78–85.
this pathway and between this and the other signaling [9] Rajkumar SV, Gupta V, Fonseca R, et al. Impact of pri-
pathways could complicate the type and depth of mary molecular cytogenetic abnormalities and risk of
response that we see in patients. In addition, the progression in smoldering multiple myeloma.
Leukemia. 2013;27:1738–1744.
heterogenous nature of MM makes this even more
[10] Keats JJ, Reiman T, Maxwell CA, et al. In multiple mye-
complicated as there could be several unique mecha- loma, t(4;14)(p16;q32) is an adverse prognostic factor
nisms that contribute to AKT pathway activation, the irrespective of FGFR3 expression. Blood. 2003;101:
type of feedback loop and the extent of cross talk 1520–1529.
with other signaling pathways. However, with more [11] Chng WJ, Gonzalez-Paz N, Price-Troska T, et al. Clinical
focused studies addressing each of these concerns and and biological significance of RAS mutations in mul-
tiple myeloma. Leukemia. 2008;22:2280–2284.
the development of more novel and potent small mol-
[12] Chng WJ, Huang GF, Chung TH, et al. Clinical and bio-
ecule inhibitors of this pathway, we remain optimistic logical implications of MYC activation: a common dif-
that such drugs will help us in controlling and possibly ference between MGUS and newly diagnosed
curing this devastating malignancy. multiple myeloma. Leukemia. 2011;25:1026–1035.
[13] Chapman MA, Lawrence MS, Keats JJ, et al. Initial gen-
ome sequencing and analysis of multiple myeloma.
Potential conflict of interest: Disclosure forms provided Nature. 2011;471:467–472.
by the authors are available with the full text of this [14] Lohr JG, Stojanov P, Carter SL, et al. Widespread gen-
article online at https://doi.org/10.1080/10428194.2017. etic heterogeneity in multiple myeloma: implications
1421760. for targeted therapy. Cancer Cell. 2014;25:91–101.
2532 V. RAMAKRISHNAN AND S. KUMAR

[15] Keats JJ, Fonseca R, Chesi M, et al. Promiscuous muta- multiple myeloma cells and required for their survival.
tions activate the noncanonical NF-kappaB pathway Cell. 2009;137:873–886.
in multiple myeloma. Cancer Cell. 2007;12:131–144. [34] Jacinto E, Loewith R, Schmidt A, et al. Mammalian
[16] Kortum KM, Mai EK, Hanafiah NH, et al. Targeted TOR complex 2 controls the actin cytoskeleton and is
sequencing of refractory myeloma reveals a high inci- rapamycin insensitive. Nat Cell Biol. 2004;6:1122–1128.
dence of mutations in CRBN and Ras pathway genes. [35] Frias MA, Thoreen CC, Jaffe JD, et al. mSin1 is neces-
Blood. 2016;128:1226–1233. sary for Akt/PKB phosphorylation, and its isoforms
[17] Kumar SK, Dispenzieri A, Lacy MQ, et al. Continued define three distinct mTORC2s. Curr Biol. 2006;16:
improvement in survival in multiple myeloma: 1865–1870.
changes in early mortality and outcomes in older [36] Pearce LR, Huang X, Boudeau J, et al. Identification of
patients. Leukemia. 2014;28:1122–1128. Protor as a novel Rictor-binding component of mTOR
[18] Di Marzo L, Desantis V, Solimando AG, et al. complex-2. Biochem J. 2007;405:513–522.
Microenvironment drug resistance in multiple mye- [37] Bracho-Valdes I, Moreno-Alvarez P, Valencia-Martinez
loma: emerging new players. Oncotarget. 2016;7: I, et al. mTORC1- and mTORC2-interacting proteins
60698–60711. keep their multifunctional partners focused. IUBMB
[19] Hideshima T, Mitsiades C, Tonon G, et al. Life. 2011;63:896–914.
Understanding multiple myeloma pathogenesis in the [38] Fernandez-Saiz V, Targosz BS, Lemeer S, et al.
bone marrow to identify new therapeutic targets. Nat SCFFbxo9 and CK2 direct the cellular response to
Rev Cancer. 2007;7:585–598. growth factor withdrawal via Tel2/Tti1 degradation
[20] Saxton RA, Sabatini DM. mTOR signaling in growth, and promote survival in multiple myeloma. Nat Cell
metabolism, and disease. Cell. 2017;168:960–976. Biol. 2013;15:72–81.
[21] Mayer IA, Arteaga CL. The PI3K/AKT pathway as a [39] Ramakrishnan V, Gomez M, Prasad V, et al. Smac
target for cancer treatment. Annu Rev Med. 2016;67: mimetic LCL161 overcomes protective ER stress
11–28. induced by obatoclax, synergistically causing cell
[22] Fruman DA, Rommel C. PI3K and cancer: lessons, chal- death in multiple myeloma. Oncotarget. 2016;7:
lenges and opportunities. Nat Rev Drug Discov. 56253–56265.
2014;13:140–156. [40] Du J, Liu S, He J, et al. MicroRNA-451 regulates stem-
[23] Ismail SI, Mahmoud IS, Msallam MM, et al. Hotspot ness of side population cells via PI3K/Akt/mTOR sig-
mutations of PIK3CA and AKT1 genes are absent in naling pathway in multiple myeloma. Oncotarget.
multiple myeloma. Leuk Res. 2010;34:824–826. 2015;6:14993–15007.
[24] Chang H, Qi XY, Claudio J, et al. Analysis of PTEN [41] Yu CF, Liu ZX, Cantley LG. ERK negatively regulates
deletions and mutations in multiple myeloma. Leuk the epidermal growth factor-mediated interaction of
Res. 2006;30:262–265. Gab1 and the phosphatidylinositol 3-kinase. J Biol
[25] Ramakrishnan V, D’Souza A. Signaling pathways and Chem. 2002;277:19382–19388.
emerging therapies in multiple myeloma. Curr [42] Zimmermann S, Moelling K. Phosphorylation and
Hematol Malig Rep. 2016;11:156–164. regulation of Raf by Akt (protein kinase B). Science.
[26] Manning BD, Cantley LC. AKT/PKB signaling: navigat- 1999;286:1741–1744.
ing downstream. Cell. 2007;129:1261–1274. [43] Guan KL, Figueroa C, Brtva TR, et al. Negative regula-
[27] Vivanco I, Sawyers CL. The phosphatidylinositol 3-kin- tion of the serine/threonine kinase B-Raf by Akt. J Biol
ase AKT pathway in human cancer. Nat Rev Cancer. Chem. 2000;275:27354–27359.
2002;2:489–501. [44] Kodaki T, Woscholski R, Hallberg B, et al. The activa-
[28] Kim DH, Sarbassov DD, Ali SM, et al. mTOR interacts tion of phosphatidylinositol 3-kinase by Ras. Curr Biol.
with raptor to form a nutrient-sensitive complex that 1994;4:798–806.
signals to the cell growth machinery. Cell [45] Rodriguez-Viciana P, Warne PH, Dhand R, et al.
2002;110:163–175. Phosphatidylinositol-3-OH kinase as a direct target of
[29] Hara K, Maruki Y, Long X, et al. Raptor, a binding part- Ras. Nature. 1994;370:527–532.
ner of target of rapamycin (TOR), mediates TOR [46] Roux PP, Ballif BA, Anjum R, et al. Tumor-promoting
action. Cell. 2002;110:177–189. phorbol esters and activated Ras inactivate the tuber-
[30] Kim DH, Sarbassov DD, Ali SM, et al. GbetaL, a posi- ous sclerosis tumor suppressor complex via p90 ribo-
tive regulator of the rapamycin-sensitive pathway somal S6 kinase. Proc Natl Acad Sci USA. 2004;101:
required for the nutrient-sensitive interaction between 13489–13494.
raptor and mTOR. Mol Cell. 2003;11:895–904. [47] Carriere A, Romeo Y, Acosta-Jaquez HA, et al. ERK1/2
[31] Sancak Y, Thoreen CC, Peterson TR, et al. PRAS40 is phosphorylate Raptor to promote Ras-dependent acti-
an insulin-regulated inhibitor of the mTORC1 protein vation of mTOR complex 1 (mTORC1). J Biol Chem.
kinase. Mol Cell. 2007;25:903–915. 2011;286:567–577.
[32] Wang L, Harris TE, Roth RA, et al. PRAS40 regulates [48] Ebi H, Costa C, Faber AC, et al. PI3K regulates MEK/ERK
mTORC1 kinase activity by functioning as a direct signaling in breast cancer via the Rac-GEF,
inhibitor of substrate binding. J Biol Chem. 2007;282: P-Rex1. Proc Natl Acad Sci USA. 2013;110:21124–21129.
20036–20044., Jr. [49] Steinbrunn T, Stuhmer T, Gattenlohner S, et al.
[33] Peterson TR, Laplante M, Thoreen CC, et al. DEPTOR is Mutated RAS and constitutively activated Akt delin-
an mTOR inhibitor frequently overexpressed in eate distinct oncogenic pathways, which
AKT PATHWAY IN MYELOMA 2533

independently contribute to multiple myeloma cell demethoxygeldanamycin has synergistic activity in


survival. Blood. 2011;117:1998–2004. multiple myeloma. Clin Cancer Res.
[50] Ramakrishnan V, Kimlinger T, Haug J, et al. Anti-mye- 2006;12:6826–6835.
loma activity of Akt inhibition is linked to the activa- [64] Baumann P, Hagemeier H, Mandl-Weber S, et al.
tion status of PI3K/Akt and MEK/ERK pathway. PLoS Myeloma cell growth inhibition is augmented by syn-
One. 2012;7:e50005. chronous inhibition of the insulin-like growth factor-1
[51] Mimura N, Hideshima T, Shimomura T, et al. Selective receptor by NVP-AEW541 and inhibition of mamma-
and potent Akt inhibition triggers anti-myeloma activ- lian target of rapamycin by Rad001. Anticancer Drugs.
ities and enhances fatal endoplasmic reticulum stress 2009;20:259–266.
induced by proteasome inhibition. Cancer Res. [65] McMillin DW, Ooi M, Delmore J, et al. Antimyeloma
2014;74:4458–4469. activity of the orally bioavailable dual phosphatidyli-
[52] Chandarlapaty S, Sawai A, Scaltriti M, et al. AKT inhib- nositol 3-kinase/mammalian target of rapamycin
ition relieves feedback suppression of receptor tyro- inhibitor NVP-BEZ235. Cancer Res. 2009;69:5835–5842.
sine kinase expression and activity. Cancer Cell. 2011; [66] Baumann P, Mandl-Weber S, Oduncu F, et al. The
19:58–71. novel orally bioavailable inhibitor of phosphoinositol-
[53] Britschgi A, Andraos R, Brinkhaus H, et al. JAK2/STAT5 3-kinase and mammalian target of rapamycin, NVP-
inhibition circumvents resistance to PI3K/mTOR block- BEZ235, inhibits growth and proliferation in multiple
ade: a rationale for cotargeting these pathways in myeloma. Exp Cell Res. 2009;315:485–497.
metastatic breast cancer. Cancer Cell. 2012;22: [67] Martin SK, Fitter S, Bong LF, et al. NVP-BEZ235, a dual
796–811. pan class I PI3 kinase and mTOR inhibitor, promotes
[54] Ramakrishnan V, Kimlinger T, Haug J, et al. TG101209, osteogenic differentiation in human mesenchymal
a novel JAK2 inhibitor, has significant in vitro activity stromal cells. J Bone Miner Res. 2010;25:2126–2137.
in multiple myeloma and displays preferential cyto- [68] Hoang B, Frost P, Shi Y, et al. Targeting TORC2 in mul-
toxicity for CD45þ myeloma cells. Am J Hematol. tiple myeloma with a new mTOR kinase inhibitor.
2010;85:675–686.
Blood. 2010;116:4560–4568.
[55] Ramakrishnan V, Ansell S, Haug J, et al. MRK003, a
[69] Maiso P, Liu Y, Morgan B, et al. Defining the role of
gamma-secretase inhibitor exhibits promising in vitro
TORC1/2 in multiple myeloma. Blood. 2011;118:
pre-clinical activity in multiple myeloma and non-
6860–6870.
Hodgkin’s lymphoma. Leukemia. 2012;26:340–348.
[70] Cirstea D, Santo L, Hideshima T, et al. Delineating the
[56] Shi Y, Gera J, Hu L, et al. Enhanced sensitivity of mul-
mTOR kinase pathway using a dual TORC1/2 inhibitor,
tiple myeloma cells containing PTEN mutations to
AZD8055, in multiple myeloma. Mol Cancer Ther.
CCI-779. Cancer Res. 2002;62:5027–5034.
2014;13:2489–2500.
[57] Shi Y, Yan H, Frost P, et al. Mammalian target of rapa-
[71] Hideshima T, Catley L, Yasui H, et al. Perifosine, an
mycin inhibitors activate the AKT kinase in multiple
oral bioactive novel alkylphospholipid, inhibits Akt
myeloma cells by up-regulating the insulin-like
and induces in vitro and in vivo cytotoxicity in human
growth factor receptor/insulin receptor substrate-1/
multiple myeloma cells. Blood. 2006;107:4053–4062.
phosphatidylinositol 3-kinase cascade. Mol Cancer
[72] Aronson LI, Davenport EL, Mirabella F, et al.
Ther. 2005;4:1533–1540.
[58] Raje N, Kumar S, Hideshima T, et al. Combination of Understanding the interplay between the proteasome
the mTOR inhibitor rapamycin and CC-5013 has syner- pathway and autophagy in response to dual PI3K/
gistic activity in multiple myeloma. Blood. 2004; mTOR inhibition in myeloma cells is essential for their
104:4188–4193. effective clinical application. Leukemia. 2013;27:
[59] Yan H, Frost P, Shi Y, et al. Mechanism by which 2397–2403.
mammalian target of rapamycin inhibitors sensitize [73] Palacios C, Martin-Perez R, Lopez-Perez AI, et al.
multiple myeloma cells to dexamethasone-induced Autophagy inhibition sensitizes multiple myeloma
apoptosis. Cancer Res. 2006;66:2305–2313. cells to 17-dimethylaminoethylamino-17-demethoxy-
[60] Ramakrishnan V, Kimlinger T, Timm M, et al. Multiple geldanamycin-induced apoptosis. Leuk Res.
mechanisms contribute to the synergistic anti-mye- 2010;34:1533–1538.
loma activity of the pan-histone deacetylase inhibitor [74] Villar VH, Nguyen TL, Teres S, et al. Escaping mTOR
LBH589 and the rapalog RAD001. Leuk Res. inhibition for cancer therapy: Tumor suppressor func-
2014;38:1358–1366. tions of mTOR. Mol Cell Oncol. 2017;4:e1297284.
[61] Simmons JK, Patel J, Michalowski A, et al. TORC1 and [75] Farag SS, Zhang S, Jansak BS, et al. Phase II trial of
class I HDAC inhibitors synergize to suppress mature temsirolimus in patients with relapsed or refractory
B cell neoplasms. Mol Oncol. 2014;8:261–272. multiple myeloma. Leuk Res. 2009;33:1475–1480.
[62] Ramakrishnan V, Timm M, Haug JL, et al. Sorafenib, a [76] Guenther A, Baumann P, Burger R, et al. Phase I/II
dual Raf kinase/vascular endothelial growth factor study with single agent everolimus (RAD001) in
receptor inhibitor has significant anti-myeloma activity patients with relapsed or refractory multiple myeloma.
and synergizes with common anti-myeloma drugs. Blood. 2009;114:3850–3850.
Oncogene. 2010;29:1190–1202. [77] Gunther A, Baumann P, Burger R, et al. Activity of
[63] Francis LK, Alsayed Y, Leleu X, et al. Combination everolimus (RAD001) in relapsed and/or refractory
mammalian target of rapamycin inhibitor rapamycin multiple myeloma: a phase I study. Haematologica.
and HSP90 inhibitor 17-allylamino-17- 2015;100:541–547.
2534 V. RAMAKRISHNAN AND S. KUMAR

[78] Yee AJ, Hari P, Marcheselli R, et al. Outcomes in relapsed/refractory multiple myeloma previously
patients with relapsed or refractory multiple myeloma treated with bortezomib: results of a multicenter
in a phase I study of everolimus in combination with phase I/II trial. JCO. 2011;29:4243–4249.
lenalidomide. Br J Haematol. 2014;166:401–409. [82] Jakubowiak AJ, Richardson PG, Zimmerman T, et al.
[79] Ghobrial IM, Weller E, Vij R, et al. Weekly bortezomib Perifosine plus lenalidomide and dexamethasone in
in combination with temsirolimus in relapsed or relapsed and relapsed/refractory multiple myeloma: a
relapsed and refractory multiple myeloma: a multi- Phase I Multiple Myeloma Research Consortium study.
centre, phase 1/2, open-label, dose-escalation study. Br J Haematol. 2012;158:472–480.
Lancet Oncol. 2011;12:263–272. [83] Spencer A, Yoon SS, Harrison SJ, et al. The novel AKT
[80] Ghobrial IM, Siegel DS, Vij R, et al. TAK-228 (formerly inhibitor afuresertib shows favorable safety, pharma-
MLN0128), an investigational oral dual TORC1/2 cokinetics, and clinical activity in multiple myeloma.
inhibitor: a phase I dose escalation study in patients Blood. 2014;124:2190–2195.
with relapsed or refractory multiple myeloma, non- [84] Tolcher AW, Patnaik A, Papadopoulos KP, et al. Phase
Hodgkin lymphoma, or Waldenstrom’s macroglobuli- I study of the MEK inhibitor trametinib in combination
nemia. Am J Hematol. 2016;91:400–405. with the AKT inhibitor afuresertib in patients with
[81] Richardson PG, Wolf J, Jakubowiak A, et al. Perifosine solid tumors and multiple myeloma. Cancer
plus bortezomib and dexamethasone in patients with Chemother Pharmacol. 2015;75:183–189.

You might also like