Lipid rafts are tightly packed, cholesterol- and sphingolipid-enriched microdomains within the plasma membrane that play important roles in many pathophysiologic processes. Rafts have been strongly implicated as master regulators of signal transduction in cancer, where raft compartmentalization can promote transmembrane receptor oligomerization, shield proteins from enzymatic degradation, and act as scaffolds to enhance intracellular signaling cascades. Cancer cells have been found to exploit these mechanisms to initiate oncogenic signaling and promote tumor progression. This review highlights the roles of lipid rafts within the metastatic cascade, specifically within tumor angiogenesis, cell adhesion, migration, epithelial-to-mesenchymal transition, and transendothelial migration. In addition, the interplay between lipid rafts and different modes of cancer cell death, including necrosis, apoptosis, and anoikis, will be described. The clinical role of lipid raft–specific proteins, caveolin and flotillin, in assessing patient prognosis and evaluating metastatic potential of various cancers will be presented. Collectively, elucidation of the complex roles of lipid rafts and raft components within the metastatic cascade may be instrumental for therapeutic discovery to curb prometastatic processes.

The presence and dynamic clustering of sphingolipid- and cholesterol-enriched “raft” domains within the plasma membrane were postulated in 1997 by Simons and Ikonen (1). This discovery added complexity to the “fluid mosaic” model presented by Singer and Nicolson, which postulated that phospholipids and membrane proteins exhibited random lateral organization within a fluid lipid bilayer (2). Lipid rafts are small arrangements enriched in specific lipids (such as saturated sphingolipids) and proteins [namely glycosylphosphatidylinositol (GPI)-anchored proteins], and the term is intended to convey the lateral heterogeneity that exists in cellular membranes (3). Elevated concentrations of cholesterol and sphingolipids containing saturated hydrophobic chains allow for more densely packed, ordered lipid arrangements, in comparison with the surrounding bilayer (4). Cholesterol, in particular, acts as both a glue and a spacer between nonpolar regions of saturated lipids, and its presence has proven vital for raft assembly and integrity. Lipid rafts have more recently been defined as transient and dynamic, both in terms of their lateral fluidity within the membrane and their constant flux of assembly/disassembly (3). The dynamic nature of rafts is highlighted by spatiotemporal changes of area and continuity between raft and nonraft regions as a result of cellular processes and stimuli. Heterogeneity also exists within lipid rafts between inner and outer leaflets of the lipid bilayer, governing biophysical properties of cellular membranes, such as curvature and rigidity, while also influencing the formation of vesicles for endo- and exocytosis (5).

The dynamic nature of lipid rafts along with their small size, short lifetime, and nonbinary characterization of “raft” and “nonraft” regions confound their experimental observation (3). The presence of heterogeneous lipid microdomains within cellular plasma membranes has been demonstrated as early as 1982, through the use of fluorescence lifetime decay of 1,6-diphenyl-1,3,5-hexatriene (6). Evidence of lipid raft microdomains was further supported by discovery of high cholesterol and glycosphingolipid regions that remained insoluble in cold, nonionic detergents (7). While detergent insolubility is still commonly used to isolate lipid rafts, validation of fluorescently labeled probes and advances in super resolution microscopy have allowed for direct visualization of raft composition, previously unseen using conventional confocal microscopy (3, 8, 9). Mass spectrometry is used to detail protein and lipid composition of raft domains, while Förster resonance energy transfer has been instrumental in probing raft presence and size (3). What remains a challenge, and what makes rafts somewhat controversial, are a lack of modalities for both live cell and in vivo observation over extended periods. Nonetheless, advances in in vitro observation and characterization reinforce their importance in cellular biology.

Lipid rafts can exist in either nonplanar (invaginated) or planar (flat) configurations (10). Caveolins are associated proteins of nonplanar lipid rafts, and are critical structural components of invaginated microdomains within the plasma membrane, known as caveolae (11). There are three known isoforms in the caveolin family; caveolin-1 and caveolin-2 are expressed ubiquitously and abundantly in epithelial cells, while caveolin-3 is highly expressed in striated and smooth muscle cells (12). On the contrary, planar rafts are smaller and remain as flat, ordered structures that are abundant in flotillin proteins (flotillin-1 and flotillin-2; refs. 1, 10). Flotillin proteins are indispensable structural components to non-caveolae planar rafts and also function as signaling platforms in the compartmentalization of membrane receptors (13).

Lipid rafts play a vital role in receptor trafficking and signal transduction, largely through their capacity to act as concentrating platforms, prompting unique interactions between clustered proteins (14). Through coalescing certain proteins, kinases, and phosphatases, while excluding others, a new lipid membrane environment is created. For example, lipid rafts protect receptors from enzymatic degradation or inactivation from non-raft enzymes. Specific receptors are predisposed to raft translocation, juxtaposing monomers to promote receptor oligomerization and supramolecular clustering, which in turn amplifies downstream signaling (15). This combination of signaling molecule concentration and exclusion of unwanted modulators make rafts master regulators of signal transduction. Within the context of cancer, these mechanisms of raft-mediated signal augmentation have been implicated in oncogenic and prometastatic signaling pathways (16). This review summarizes the roles that lipid rafts play within cancer metastasis, cell death, and raft-associated clinical biomarkers.

The metastatic dissemination of cancer cells from the primary tumor to distant organs is a process that few cells survive (17). Successful metastatic colonization requires the following: creation of tumor neovasculature to support nutrient and oxygen demand, enhanced cellular motility through an epithelial-to-mesenchymal transition (EMT), directed migration and transendothelial migration (TEM) into the vasculature, survival of aberrant shear conditions in circulation, endothelial rolling and extravasation, and a phenotypic switch back to a proliferative state. Despite its inefficiencies, metastasis is responsible for approximately 90% of all cancer-related deaths (18). Understanding the complex protein interactions that allow for successful dissemination of specific subpopulations of cells is critical for discovery and development of antimetastatic therapies. Herein, the roles of lipid rafts within the steps of cancer metastasis will be discussed (Table 1A) and illustrated (Fig. 1).

Table 1A.

Lipid rafts in metastatic stages, including angiogenesis, EMT, cell migration, TEM, and cell adhesion, along with mechanisms of cell death, including necrosis, apoptosis, and anoikis.

Important proteins/pathwayLipid rafts effect on stageCell linesLipid raft treatmentReferences
Angiogenesis TEK, RhoA Promote MDA-MB-231, MDA-MB-468, ZR 751, HUVECs, HMEC Cerivastatin, mβCD, nystatin, and filipin III (inhibitors) (19–21) 
 VEGF/VEGFR2 Promote PC-3, LNCaP, LLC-1, HT-29, SW-480, HUVECs CAV1 siRNA (24–27) 
 Caveolin-1, CD82, CD24/Hsp90/STAT3     
 Exosomal miR-205 Promote HO-8910, SKOV-3 Filipin III, simvastatin (inhibitors) (29) 
EMT TGFβ/MAPK Promote HaCaT, NMuMG Nystatin (32) 
 ERK, p38     
 Integrin-β3 Promote A549, PC-9 Simvastatin (30) 
 pFAK/pAkt/pERK/TGF-β     
 HFG/c-Met Promote MCF7, MDA-MB-453, MDA-MB-231, BT-20 Osthole (inhibitor) (34) 
 pAkt, mTOR     
 Podoplanin Promote MDCK type II mβCD (35) 
 ROCK/ezrin/radixin/moesin     
 Caveolin-1/c-MET Promote T24, UMUC3, HT1376, 5637, MGC-803, SGC-7901, BGC-823 CAV1 siRNA, nystatin (31, 36) 
 CXCL12/CXCR4, Slug, PI3K/Akt     
 Flotillin-2 Promote GES-1, SGC-7901, NCI-N87, MGC-803 Flotillin-2 siRNA (37) 
 TGFβ/vimentin/N-cadherin     
 Snail, Slug Inhibit MDA-MB-231 DHA (lipid raft stabilizer) (38) 
Migration Integrin-β1/MMP-9/uPAR Promote P29, MDA-MB-231, ZR-751, HMEC mβCD, nystatin (21, 44) 
 Src, FAK, Cav, Akt, ERK     
 EGFR Promote MDA-MB-231, MCF7, T47D mβCD (50) 
 Akt/PKB     
 CD133/Caveolin-1 Promote MIA PaCa-2, Panc-1, SU.86.86 Lovastatin (inhibitor), CAV1 siRNA (54) 
 FAK, NF-κB     
 Flotillin-2 Promote MDA-MB-231, MCF7 Flotillin-2 shRNA (52, 53) 
 PI3K/Akt     
 SK3/Orai1 Promote MDA-MB-435 Alkyl-lipid ohmline (inhibitor) (42) 
 Calpain/talin     
 Squalene synthase/TNFR1 Promote A549, CL1–5 mβCD (45) 
 MMP1/NF-κB     
 CXCL12/CXCR4 Promote C4-2B, LNCaP, PC3, MGC-803, SGC-7901, BGC-823 Nystatin, CAV1 siRNA (31, 48, 49) 
 Caveolin-1/C-met, EGFR, Gαi/HER2/Src, PI4KIIIα     
 Artificial crosslinking of GPI-anchored placental alkaline phosphatase Promote A375, HeLa Ru-complex–based trefoil molecule (41) 
 TRPM8/AR Promote PC3, LNCaP  (51) 
 FAK     
TEM C1QBP/CD44vg Promote H MIA-PaCa‐2, PANC‐1, SW1990, Capan‐1, BxPC‐3  (58) 
 IGF-1R/PI3K/MAPK     
 Caveolin-1/MT1-MMP Promote MDA-MB-231, T47D, BT549, Hs578T, MDA-MB-453, SK-BR-3 mβCD, CAV1 siRNA, nystatin (64, 65) 
 PI3K/Akt/mTOR     
 Caveolin-1/FAPα Promote CAFs  (63) 
 Podoplanin Promote HaCaT, SCC29, A253, Fadu, HN30, SCC13, HN5 mβCD (66) 
 RhoC-GTPase/ROCK/LIMK/Cofilin     
Adhesion Fibronectin and vitronectin adhesion Promote MDA-MB-231, MDA-MB-468 mβCD (20) 
 Integrin-β1/focal adhesion complex proteins Promote MDA-MB-231, HeLa, HSC5, HepG2, HCT-116 mβCD, simvastatin, emodin, gambogic acid (inhibitors) (40, 85, 86) 
 CD24 and CD44/Src/Integrin-β1 Promote U251, H1299, H23, H460, A569, SHP-77, AZ521, HT-29, MTLy, RKO, HCT-116 mβCD, Filipin, Simvastatin (80–82, 84) 
 FAK, paxillin     
 CD44 Inhibit MCF10a, MDA-MB-231  (87) 
 Ezrin     
 SFK Promote MCF7, MDA-MB-231 LRT-SIFP (inhibits the SFK activity in LRs) (83) 
 Integrin-α5β1/LFA-1/β3 Promote Jurkat, F111, P-815 mβCD, mal-βCD (inhibitor) (70, 72, 73) 
Necrosis MLKL Promote HEK293T, L929, Jurkat, HeLa, CHO, SKOV3, TOV112D, A2780, A2780CP, PE01, PE04  (95–97) 
 TNFa/RIPK1/RIPK3     
Anoikis Integrin/FAK Inhibit A431, PZ-HPV7, MCF10A, MCF7, MDA-MB-231, PC-3, LNCaP Simvastatin, mβCD (113, 114) 
 Bcl-xL, Akt, Src     
 HIF1α Inhibit A431, HeLa mβCD (115) 
 EGFR/Akt/mTOR     
Apoptosis DR4/DR5 Promote HT29, HCT116, SW480, SW620, H460, A549, A549, H1792, H596, CEM, HD-MyZ, I-83, JMV-3, NALM-6, BJAB Nystatin (102–106) 
 Fas/CD95, FADD, procaspase-8/10, JNK, Bid, Caspase-3 Promote HL-60, Jurkat, MM1S, MM144, U266 mβCD, filipin (100, 101) 
 Akt Inhibit NIH3T3 mβCD (110) 
Important proteins/pathwayLipid rafts effect on stageCell linesLipid raft treatmentReferences
Angiogenesis TEK, RhoA Promote MDA-MB-231, MDA-MB-468, ZR 751, HUVECs, HMEC Cerivastatin, mβCD, nystatin, and filipin III (inhibitors) (19–21) 
 VEGF/VEGFR2 Promote PC-3, LNCaP, LLC-1, HT-29, SW-480, HUVECs CAV1 siRNA (24–27) 
 Caveolin-1, CD82, CD24/Hsp90/STAT3     
 Exosomal miR-205 Promote HO-8910, SKOV-3 Filipin III, simvastatin (inhibitors) (29) 
EMT TGFβ/MAPK Promote HaCaT, NMuMG Nystatin (32) 
 ERK, p38     
 Integrin-β3 Promote A549, PC-9 Simvastatin (30) 
 pFAK/pAkt/pERK/TGF-β     
 HFG/c-Met Promote MCF7, MDA-MB-453, MDA-MB-231, BT-20 Osthole (inhibitor) (34) 
 pAkt, mTOR     
 Podoplanin Promote MDCK type II mβCD (35) 
 ROCK/ezrin/radixin/moesin     
 Caveolin-1/c-MET Promote T24, UMUC3, HT1376, 5637, MGC-803, SGC-7901, BGC-823 CAV1 siRNA, nystatin (31, 36) 
 CXCL12/CXCR4, Slug, PI3K/Akt     
 Flotillin-2 Promote GES-1, SGC-7901, NCI-N87, MGC-803 Flotillin-2 siRNA (37) 
 TGFβ/vimentin/N-cadherin     
 Snail, Slug Inhibit MDA-MB-231 DHA (lipid raft stabilizer) (38) 
Migration Integrin-β1/MMP-9/uPAR Promote P29, MDA-MB-231, ZR-751, HMEC mβCD, nystatin (21, 44) 
 Src, FAK, Cav, Akt, ERK     
 EGFR Promote MDA-MB-231, MCF7, T47D mβCD (50) 
 Akt/PKB     
 CD133/Caveolin-1 Promote MIA PaCa-2, Panc-1, SU.86.86 Lovastatin (inhibitor), CAV1 siRNA (54) 
 FAK, NF-κB     
 Flotillin-2 Promote MDA-MB-231, MCF7 Flotillin-2 shRNA (52, 53) 
 PI3K/Akt     
 SK3/Orai1 Promote MDA-MB-435 Alkyl-lipid ohmline (inhibitor) (42) 
 Calpain/talin     
 Squalene synthase/TNFR1 Promote A549, CL1–5 mβCD (45) 
 MMP1/NF-κB     
 CXCL12/CXCR4 Promote C4-2B, LNCaP, PC3, MGC-803, SGC-7901, BGC-823 Nystatin, CAV1 siRNA (31, 48, 49) 
 Caveolin-1/C-met, EGFR, Gαi/HER2/Src, PI4KIIIα     
 Artificial crosslinking of GPI-anchored placental alkaline phosphatase Promote A375, HeLa Ru-complex–based trefoil molecule (41) 
 TRPM8/AR Promote PC3, LNCaP  (51) 
 FAK     
TEM C1QBP/CD44vg Promote H MIA-PaCa‐2, PANC‐1, SW1990, Capan‐1, BxPC‐3  (58) 
 IGF-1R/PI3K/MAPK     
 Caveolin-1/MT1-MMP Promote MDA-MB-231, T47D, BT549, Hs578T, MDA-MB-453, SK-BR-3 mβCD, CAV1 siRNA, nystatin (64, 65) 
 PI3K/Akt/mTOR     
 Caveolin-1/FAPα Promote CAFs  (63) 
 Podoplanin Promote HaCaT, SCC29, A253, Fadu, HN30, SCC13, HN5 mβCD (66) 
 RhoC-GTPase/ROCK/LIMK/Cofilin     
Adhesion Fibronectin and vitronectin adhesion Promote MDA-MB-231, MDA-MB-468 mβCD (20) 
 Integrin-β1/focal adhesion complex proteins Promote MDA-MB-231, HeLa, HSC5, HepG2, HCT-116 mβCD, simvastatin, emodin, gambogic acid (inhibitors) (40, 85, 86) 
 CD24 and CD44/Src/Integrin-β1 Promote U251, H1299, H23, H460, A569, SHP-77, AZ521, HT-29, MTLy, RKO, HCT-116 mβCD, Filipin, Simvastatin (80–82, 84) 
 FAK, paxillin     
 CD44 Inhibit MCF10a, MDA-MB-231  (87) 
 Ezrin     
 SFK Promote MCF7, MDA-MB-231 LRT-SIFP (inhibits the SFK activity in LRs) (83) 
 Integrin-α5β1/LFA-1/β3 Promote Jurkat, F111, P-815 mβCD, mal-βCD (inhibitor) (70, 72, 73) 
Necrosis MLKL Promote HEK293T, L929, Jurkat, HeLa, CHO, SKOV3, TOV112D, A2780, A2780CP, PE01, PE04  (95–97) 
 TNFa/RIPK1/RIPK3     
Anoikis Integrin/FAK Inhibit A431, PZ-HPV7, MCF10A, MCF7, MDA-MB-231, PC-3, LNCaP Simvastatin, mβCD (113, 114) 
 Bcl-xL, Akt, Src     
 HIF1α Inhibit A431, HeLa mβCD (115) 
 EGFR/Akt/mTOR     
Apoptosis DR4/DR5 Promote HT29, HCT116, SW480, SW620, H460, A549, A549, H1792, H596, CEM, HD-MyZ, I-83, JMV-3, NALM-6, BJAB Nystatin (102–106) 
 Fas/CD95, FADD, procaspase-8/10, JNK, Bid, Caspase-3 Promote HL-60, Jurkat, MM1S, MM144, U266 mβCD, filipin (100, 101) 
 Akt Inhibit NIH3T3 mβCD (110) 

Note: Proteins in lipid rafts are bolded and associated pathways/proteins are italicized.

Figure 1.

Lipid rafts play an important role in cancer cell metastasis and cell death. Angiogenesis: CD24 raft colocalization promotes VEGF secretion of cancer cells, while VEGFR2 raft colocalization enhances angiogenesis upon VEGF activation (24, 27). EMT: TGFβ receptor is reliant on localization with rafts to promote downstream EMT signaling pathways (32). Migration: Raft colocalization promotes MMP-9 activation and CXCR4-mediated cell migration (31, 44). TEM: Lipid rafts are enriched at invadopodia where caveolin-1 colocalizes with MT1-MMP to promote TEM (64, 65). Cell death: Integrin–ECM detachment leads to FAK deactivation and the anoikis response (112). Adhesion: colocalization of CD44 and β1 integrin enhance matrix-dependent cell adhesion to hyaluronic acid and fibronectin, respectively (20, 82).

Figure 1.

Lipid rafts play an important role in cancer cell metastasis and cell death. Angiogenesis: CD24 raft colocalization promotes VEGF secretion of cancer cells, while VEGFR2 raft colocalization enhances angiogenesis upon VEGF activation (24, 27). EMT: TGFβ receptor is reliant on localization with rafts to promote downstream EMT signaling pathways (32). Migration: Raft colocalization promotes MMP-9 activation and CXCR4-mediated cell migration (31, 44). TEM: Lipid rafts are enriched at invadopodia where caveolin-1 colocalizes with MT1-MMP to promote TEM (64, 65). Cell death: Integrin–ECM detachment leads to FAK deactivation and the anoikis response (112). Adhesion: colocalization of CD44 and β1 integrin enhance matrix-dependent cell adhesion to hyaluronic acid and fibronectin, respectively (20, 82).

Close modal

Angiogenesis

Lipid rafts play an important role in angiogenic signaling, as demonstrated in studies of raft antagonization. One study found that treatment with cerivastatin, an inhibitor of cholesterol biosynthesis, which disrupts lipid rafts, attenuated angiogenesis by inhibiting endothelial cell migration and capillary formation (19). Another study found that raft disruption in triple-negative breast cancer (TNBC) cells via the cholesterol-depleting agent, methyl-beta-cyclodextrin (mβCD), inhibited angiogenic signaling through the suppression of proangiogenic markers, namely tyrosine protein kinase receptor (20). Likewise, conditioned media from mβCD-treated cancer cells decreased endothelial branching and capillary structures, demonstrating the obligatory roles of lipid rafts in tumor angiogenesis (21). Lipid rafts have also been found to mediate and activate heparinase-induced protein kinase B (Akt/PKB) phosphorylation, a proangiogenic response in hypervascularized tumors (22).

The VEGF/VEGFR2 axis is a proangiogenic signaling pathway employed by cancer cells via enhanced VEGF secretion (23). Lipid rafts have proven to be important in endothelial cell VEGFR2 functionality. For example, one study found that affecting endothelial cholesterol homeostasis via liver X receptor activation impaired VEGFR2 compartmentalization in lipid rafts, leading to defective VEGFR2 phosphorylation and downstream signaling upon VEGF-A stimulation (24). Another study found that lipid raft marker caveolin-1 secreted by metastatic prostate cancer cells, potentiated angiogenic signaling though colocalization and autophosphorylation of endothelial cell VEGFR2 (25). Moreover, a separate study found that by inhibiting CD82 internalization and distribution into lipid rafts, VEGFR2 phosphorylation was reduced, attenuating angiogenic signaling (26).

Lipid rafts not only play a role in receptor signaling, but in cancer cell–secreted VEGF as well. Lipid raft–localized Hsp90 was found to stabilize CD24, necessary for STAT3 activation and VEGF angiogenic signaling in colorectal cancer cells (27). Lipid rafts are also involved in exosome uptake, enabling noncoding miRNA communication between cancer cells, the endothelium, and stroma (28). One study found that endothelial cell internalization of ovarian cancer–derived exosomal miR-205 occurred in a lipid raft–dependent manner. Inhibition of lipid raft–mediated endocytosis curbed miR-205 uptake, thereby inhibiting angiogenesis (29). These studies demonstrate specific instances of raft-promoted tumor angiogenesis, making cholesterol modulation an appealing target to prevent tumor vascularization.

EMT

Lipid raft disruption has been shown to attenuate EMT in cancer. One study found that disrupting lipid rafts with simvastatin suppressed integrin-β3/FAK signaling, reversing EMT and EMT-associated paclitaxel resistance in non–small cell lung cancer (NSCLC) cells (30). Another study found that nystatin, a cholesterol sequestering agent, suppressed EMT markers in gastric cancer cells (31). They further demonstrated that CXCL12/CXCR4-induced mesenchymal-to-epithelial transition factor (c-MET) activation and EMT are dependent on lipid raft caveolin-1 signaling. Nystatin treatment also attenuated EMT by disrupting TGFβ signaling (32), which is strongly implicated in EMT and enhanced cancer cell motility (33). Cholesterol was found to be essential for the TGFβ receptor activation of MAPK and subsequent EMT induction (32). This TGFβ–EMT synergy was contingent upon TGFβ receptor localization into lipid rafts for efficient downstream signaling. Osthole, another lipogenic modulator, inhibits EMT via lipid raft depletion. A study found that osthole suppressed the hepatocyte growth factor/c-Met signaling pathway in breast cancer cells, abrogating a mesenchymal phenotype through downregulation of vimentin and upregulation of E-cadherin (34). Meanwhile, another study found that localization of the EMT-promoting glycoprotein, podoplanin, into lipid raft domains was necessary for podoplanin-mediated EMT and cell migration (35). Treatment with mβCD impaired podoplanin lipid raft compartmentalization and impeded EMT.

The flotillin and caveolin families of proteins are also important drivers of EMT. One study found that upregulation of caveolin-1 is associated with enhanced EMT and aggressive, metastatic bladder cancer (36). Caveolin-1 enhanced EMT by upregulating Slug, an EMT promoting transcription factor, through PI3K/AKT signaling. Knockdown of caveolin-1 reduced Slug expression, subsequently inhibiting EMT. Likewise, flotillin-2 is upregulated in gastric cancer cells and is required for TGFβ-induced EMT (37). Inhibition of flotillin-2 reduced EMT markers, vimentin and N-cadherin.

While much work has shown that disrupting lipid rafts prevents EMT signaling, other research shows that lipid raft destabilization is required for EMT. One study found that lipid raft stability decreases during EMT in breast cancer cells (38). Stabilizing lipid rafts with docosahexaenoic acid (DHA) abrogated EMT hallmarks, prevented mesenchymal signaling, and inhibited metastasis. This demonstrates the complex interplay between rafts and EMT, warranting further validation before clinical implementation of raft destabilizers.

Migration

Because lipid rafts are closely tied to cytoskeletal components and receptor signaling, many studies have looked at their role within focal adhesions and cancer cell migration (39). For example, disrupting lipid rafts in human melanoma decreased migration velocity and inhibited lamellipodia formation by inducing the formation of actin stress fibers, preventing the disassembly of focal adhesions (40). One interesting study designed a ruthenium complex–based peptidic molecule that self-assembles into nanofibrils on lipid rafts (41). The nanofibrils cross-linked lipid rafts, causing them to aggregate into large chained structures. This raft constriction caused focal adhesion suppression and decreased cell migration. Another study found that the potassium channel, SK3, associates with calcium channel, Orai1, in lipid rafts to upregulate calcium influx and promote cell migration and bone metastasis (42). Treating with the alkyl-lipid, ohmline, translocated the SK3–Orai1 complex out of lipid rafts, impairing calcium entry and attenuating cell migration.

Matrix metallopeptidase proteins (MMP) play a critical role in extracellular matrix (ECM) degradation during cancer cell migration (43). One study found that enhanced MMP-9 localization into lipid rafts augmented cell migration and metastatic potential in mouse Lewis lung cancer (44). Disrupting rafts with mβCD significantly decreased MMP-9 secretion and suppressed invasiveness of these highly metastatic cells. Similarly, another study found that mβCD treatment reduced colocalization of the GPI-anchored membrane protein, uPAR, and MMP-9 in lipid rafts, decreasing migration of breast cancer cells (21). MMP-1 is also important in promoting cancer cell migration. A study found that squalene synthase was upregulated in metastatic lung cancers and caused lipid raft enrichment of TNF receptor-1 (TNFR1), enhancing NF-κB activation and leading to MMP-1 upregulation (45). Treatment with mβCD inhibited TNFR1–lipid raft colocalization, abrogating promigratory signaling and reducing the metastatic potential of lung cancer cells.

Lipid rafts also influence chemotactic signaling of directed cell migration. The CXCL12/CXCR4 chemokine axis has been extensively studied for its role in driving cancer cell migration (46, 47). In metastatic prostate cancer, this signaling axis was found to transactivate EGFR, HER2, and Src selectively within lipid raft microdomains (48). CXCR4 has been shown to colocalize with phosphatidylinositol 4-kinase IIIα (PI4KIIIα) within lipid rafts to promote CXCL12-stimulated cell invasion (49). Likewise, CXCL12-induced c-MET activation and cell migration were found to be dependent on lipid raft protein, caveolin-1, and inhibition of rafts with nystatin decreased activation of the CXCL12/CXCR4 axis (31). Lipid rafts are also essential for androgen receptor- and EGFR-mediated cell migration (50). EGFR was found to colocalize into lipid rafts, while mβCD raft disruption inhibited EGF-induced chemotaxis and actin polymerization in breast cancer cells. In prostate cancer cells, low testosterone caused accumulation of transient receptor potential melastatin 8 (TRPM8) with androgen receptors in lipid rafts, thereby inhibiting TRPM8 and promoting cell migration (51).

Flotillin and caveolin are also implicated in driving cell migration. By reducing levels of flotillin-2 in breast cancer cells, tumor volume, metastatic capability, and proliferation decreased by inhibiting PI3K/Akt signaling (52, 53). In addition, CD133, an oncogenic cancer stem cell (CSC) marker, has been shown to colocalize with caveolin-1 in lipid rafts to increase invasiveness and chemoresistance in pancreatic tumor–initiating cells (54).

TEM

Metastatic dissemination relies on the cell's ability to enter into (intravasation) and exit from (extravasation) the vasculature via TEM (55). The MUC1/intercellular adhesion molecule-1 (ICAM1) interaction activates a cascade through which physical barriers for TEM are abrogated (56). MUC1 proteins, found to be concentrated in lipid rafts, bind to ICAM1 on endothelial cells to facilitate TEM (57). In pancreatic cancer cells, one study found that IGF1 induces CD44/C1QBP complex formation in lipid rafts, activating PI3K/MAPK signaling pathways and promoting TEM. Knockdown of C1QBP inhibited complex formation in rafts, decreasing TEM (58).

Multiple studies have established that invadopodia formation is required for successful intravasation and extravasation (59, 60). For example, one study found that invadopodia formation through N-WASP–mediated actin cytoskeleton reorganization is required for cancer cell intravasation (61). Another study found that cells extend invadopodia through the endothelium prior to extravasation; by inhibiting invadopodia formation, extravasation and metastatic tumor formations were decreased (62). There is strong evidence that invadopodia formation in cancer cells relies on lipid raft enrichment. Proteases, FAPα and MT1-MMP, were found to colocalize with caveolin-1 in lipid rafts, recruiting invadopodia in cancer-associated fibroblasts (CAF) and breast cancer cells, respectively (63, 64). Disruption of lipid rafts via mβCD and CAV1 gene silencing impeded MT1-MMP activation and suppressed invadopodia formation (64). Similarly, another study found that caveolin-1 activates MT1-MMP in invadopodia through the PI3K/Akt/mTOR pathway under low shear stress (65). Treatment with mβCD inhibited MT1-MMP expression and prevented invadopodia formation, while CAV1 silencing curbed metastatic formation in animal models. An additional study revealed that podoplanin is recruited into invadopodia via lipid rafts and is essential for invadopodia stability by controlling the ROCK–LIMK–Cofilin pathway (66). Taken together, these studies show that lipid rafts are important facilitators of TEM mechanisms and raft perturbation may be a viable therapeutic strategy to curb intravasation and extravasation.

Cancer cell adhesion

The likelihood of cancer cells surviving the disadvantageous stresses within the metastatic cascade is contingent upon their interactions with the ECM, fibroblasts, bloodborne cells, and the endothelium (67). Cholesterol and sphingolipids, two integral components of lipid rafts, are necessary for the ECM adhesion of cancer cells (68, 69). Elevated cholesterol induces redistribution of integrins, resulting in increased cell attachment to fibronectin (70). Conversely, membrane sphingolipid depletion prevents binding to fibronectin (71). One study found that while lipid raft levels did not contribute to de novo synthesis of integrin, the depletion of lipid rafts with 6-O-α-maltosyl-β-cyclodextrin (mal-βCD) demonstrated that lipid raft levels were directly correlated with integrin activation and fibronectin adhesion (72). A different study obtained similar results with human T cells. In both primary human T cells and Jurkat lymphoma cells, mβCD-induced raft depletion resulted in decreased integrin-α5β1 and -αLβ2 (LFA-1)-mediated adhesion (73). These studies reinforce the role that lipid rafts play in contributing to integrin-mediated ECM adhesion.

CD44 has been implicated in cancer progression and metastasis as a dynamic regulator of cell migration and adhesion, and its roles include initiating circulating tumor cell (CTC) adhesion and rolling on the endothelium (74–76). Similarly, integrins, particularly integrins of the β1 subtype, are critical for cancer cell adhesion and vasculature survival in transit (77–79). The translocation of integrins and adhesion-related cluster of differentiation molecules to lipid rafts is critical for adhesion-related maintenance of cell migration. One study found that raft disruption of TNBC cells via mβCD treatment significantly decreased cell adhesion on fibronectin- and vitronectin-coated substrates (20). Likewise, cholesterol depletion caused CD44 shedding from lipid rafts in cancer cells, suppressing adhesion, migration, and endothelial cell rolling (80, 81). CD44 is known to interact with C1QBP in rafts to activate PI3K/MAPK downstream, a signaling pathway that also promotes cell adhesion and other prometastatic phenotypes (58). In addition, CD44 clustering in lipid rafts was found to activate Src family kinases (SFK), enriching β1 integrins into lipid rafts to promote cell adherence and matrix-derived survival (82). By inhibiting SFK activity in lipid rafts, cancer cell adhesion of breast cancer cells was inhibited (83). Similarly, CD24 was found to interact with, and promote c-Src translocation into lipid rafts, enhancing formation of focal adhesions, integrin-mediated adhesion, and cell spreading (84). Using mβCD, emodin, and gambogic acid, studies have shown that blocking focal adhesion complex protein localization into lipid rafts inhibits tumor cell adhesion (85, 86).

While many studies support the promigratory role of lipid raft–colocalized CD44, its role remains controversial. For example, one study demonstrated that enhanced palmitoylation of CD44 drives colocalization with rafts, limiting associations with its cytoskeletal linker binding partner, ezrin, to suppress migration in invasive breast cancer subtypes (87). Indeed, raft affinity of CD44 is largely regulated by one of two mechanisms: palmitoylation, which enhances affinity and reduces binding to cytoskeletal linkers, and phosphatidylinositol 4,5-bisphosphate (PIP2) membrane concentration, which decreases raft affinity, thereby accelerating formation of the CD44–adaptor complex (88). This confounding role of CD44 may be explained by the dynamic nature of protein–raft colocalization, that is, while palmitoylated CD44 may exist within rafts in an “inactive” state, rapid depalmitoylation may induce raft dissociation and subsequently promote cell adhesion. Meanwhile, CD44 shredding from rafts as a result of cholesterol depletion prevents the possibility of CD44 dissociation to “nonraft” PIP2 localized regions, thereby abrogating CD44-mediated cell adherence.

Cell death is a vital function that occurs naturally in the body, one that mediates the removal of damaged or infected cells and maintains tissue homeostasis (89). Mechanisms of cell death are especially important in the body's response to cancer; most cells that develop DNA abnormalities and cell checkpoint mutations undergo programmed cell death before they can proliferate further (90). Immunity to this process is one of the hallmarks of cancer, resulting in uncontrollable division and metastasis (91). The roles of lipid rafts in various types of cell death, including necrosis, apoptosis, and anoikis, have been studied extensively in recent years (Table 1A).

Necrosis

Necrosis is a form of unprogrammed cell death that occurs in response to trauma and other stress-inducing stimuli, resulting in an inflammatory cell death response (92). More recently, it has been suggested that necrotic cell death proceeds in a programmed manner similar to apoptosis (93). This pathway, termed “necroptosis,” acts through a caspase-independent signaling cascade. Several studies have evaluated the role of lipid rafts in this necroptotic pathway.

Mixed lineage kinase domain-like pseudokinase (MLKL) has been identified as a downstream protein in the TNFα-induced necroptosis signaling cascade (94). One study demonstrated that oligomerization of MLKL proteins and translocation into lipid rafts are necessary for necroptosis in murine fibrosarcoma cells (95). Translocation of the oligomerized MLKL complex into lipid rafts supported sodium influx, increased osmotic pressure, and cell rupture. Similarly, another study demonstrated MLKL-induced membrane permeabilization of PIP-containing liposomes (96). Through selective mutation, it was discovered that binding of positive amino acids on the oligomerized MLKL complex to PIPs allows for recruitment to lipid rafts. Inhibiting PI5P and PIP2 led to decreased necroptosis in Jurkat and murine fibrosarcoma cells. A subsequent study evaluated the effect of ceramide nanoliposomes (CNL) on MLKL expression, demonstrating that CNLs strongly promoted MLKL activation–driven necroptosis, but not apoptosis in ovarian cancer cells (97). These results suggest that CNLs form lipid raft mimetics, which promote MLKL translocation and necroptosis, supporting the role of lipid rafts in necroptosis. Given that many cancer cells display resistance to apoptosis, therapeutic targeting and activation of the necroptotic pathway present an exploitable alternative to induce cell death.

Apoptosis

Apoptosis, is a noninflammatory, programmed cell death pathway (89, 98). External activation of death receptor in the TNF superfamily results in the recruitment of the death-inducing signaling complex, caspase activation, DNA fragmentation, and cell death (99). The majority of lipid raft–apoptosis synergism studies are in relation to the colocalization of CD95 (Fas) and death receptors 4/5 into rafts, forming clusters of apoptotic signaling molecule-enriched rafts to enhance apoptotic signaling (100–105). Chemotherapeutic agents, such as perifosine, and lipid raft agonists, such as resveratrol, have been shown to promote death receptor translocation into rafts, warranting further investigation into combination treatments of chemotherapeutic raft synergism with death-inducing ligands (101, 106, 107). For additional information, studies of raft-mediated death ligand signaling have been extensively covered in recent reviews (16, 108, 109).

Apart from death receptor signaling, some studies have conversely implicated rafts as being prosurvival, apoptotic regulators. For example, one study demonstrated that mβCD-induced raft disruption resulted in G2–M-phase arrest and eventual apoptosis in breast cancer cells (20). Likewise, protein kinase B (Akt), an important prosurvival protein, was shown to localize into lipid rafts (110). Lipid raft disruption using mβCD led to a decrease of Akt activity and increased apoptosis in mouse fibroblast cells.

Anoikis

Anoikis is a specialized type of apoptosis that is activated upon cell detachment, and developed resistance to anoikis is an essential step in cancer metastasis (111, 112). The loss of integrin attachment to matrix results in an inactivation of apoptosis inhibitor, FAK, leading to caspase activation. Because lipid rafts facilitate integrin interactions and cell adhesion, depletion of lipid rafts has also been shown to directly result in anoikis-like death. When treated with cholesterol-inhibiting agents, human breast, prostate, and epidermoid carcinoma cells showed decreased lipid raft formation, Bcl-2 downregulation, caspase-3 activation, and Akt downregulation, inducing anoikis-like apoptosis (113). Similarly, another study demonstrated that simvastatin-induced cholesterol depletion resulted in raft internalization and FAK downregulation, resulting in cell detachment, caspase-3 activation, and anoikis (114).

The lipid raft association of hypoxia inducible factor 1 (HIF1) in cell attachment has also been studied. HIF1α is produced under hypoxic conditions and promotes cell survival. Interestingly, hypoxic conditions have been shown to correlate with decreased lipid rafts, suggesting that HIF1α influences lipid raft production. Epidermoid carcinoma cells treated with mβCD under normoxic conditions demonstrated upregulated HIF1α, suggesting that the cells underwent HIF1α-mediated lipid raft production in response to cholesterol depletion (115). Silencing of HIF1α led to accelerated cell detachment and anoikis. These studies demonstrate the complexities of lipid raft involvement in mechanisms of cell death. Exploiting anoikis-like apoptosis mechanisms through lipid raft augmentation may be a viable method to prevent cancer cell survival upon detachment.

Apart from their roles in cancer metastasis and cell death, components of lipid rafts, namely caveolin and flotillin, have received much attention as clinical biomarkers. There are confounding oncogenic and tumor-suppressing roles of these protein families surrounding different aspects of tumor growth and metastasis observed clinically. This section aims to elucidate the seemingly contradictory roles of caveolins and flotillins in metastatic progression and patient prognosis for a range of cancers. The prognostic roles of these proteins are summarized in Table 1B.

Table 1B.

The role of flotillin and caveolin families of proteins in metastatic progression and patient prognosis.

Protein familyCancer typeSummaryProteinPrognosisReferences
Flotillin Breast cancer ↑ flotillin-2 is significantly correlated with clinical stage, metastasis, and shorter overall survival Flotillin-2 Poor (119) 
 Cervical cancer ↑ flotillin-1 and flotillin-2 corelates with pelvic lymph node metastasis, clinical stage, tumor differentiation, and poor overall survival Flotillin-1 and -2 Poor (120, 121) 
 Colorectal cancer ↑ flotillin-1 and flotillin-2 are associated with tumor volume, depth of invasion, lymph node metastasis, distant metastasis, increased proliferation, and poor survival Flotillin-1 and -2 Poor (125, 126) 
 Liver (hepatocellular and intrahepatic cholangio carcinoma) ↑ flotillin-1 and flotillin-2 in cancerous tissue and positively correlated with tumor size, clinical stage, vascular invasion, lymph node metastasis, and relapse. Flotillin-1 and -2 Poor (123, 124) 
 Nasopharyngeal carcinoma ↑ flotillin-2 promotes tumor progression and is positively associated with a metastatic phenotype and sorter overall survival Flotillin-2 Poor (128, 129) 
 Neuroblastoma ↓ flotillin-1 correlated with poor prognosis and mRNA expression inversely correlate with clinical malignancy grade Flotillin-1 Favorable (130) 
 Small-cell lung cancer ↑ flotillin-1 is highly expressed in small-cell lung cancer and strongly correlates with clinical stage, distant metastasis, and poor survival Flotillin-1 Poor (127) 
 Various ↑ flotillin-1 and flotillin-2 are associated with shorter overall survival, decreased disease-free survival, and increased lymph node metastasis Flotillin-1 and -2 Poor (117, 118) 
 Vulvar squamous cell carcinoma ↑ flotillin-1 predicts poor overall and progressive free survival and is an oncogenic facilitator of inguinal/femoral lymph node metastasis Flotillin-1 Poor (122) 
Caveolin Bladder cancer ↑ caveolin-1 in cancerous tissues and high-grade tumors Caveolin-1 Poor (132, 133) 
 Breast cancer ↑ stromal caveolin-1 in CAFs correlated with increased 5-year survival, low histologic grade while absence or ↓ stromal caveolin-1 increased risk of recurrence and predicted lymph node metastasis Caveolin-1 (stromal and epithelial) Favorable (stromal) (142–145) 
  ↑ epithelial caveolin-1 correlates with high histologic grade, lack of hormone receptors, and decreased survival  Poor (tumoral)  
  Tumor + stromal cancer subtypes had poor survival    
 Breast cancer (brain metastases) ↑ caveolin-1 negatively regulates Stat3, inhibiting brain metastasis from breast cancer Caveolin-1 Favorable (146) 
 Colorectal cancer (liver metastases) ↓ stromal caveolin-1 is associated with decreased disease-free and overall survival after hepatectomy Caveolin-1 (stromal) Favorable (137) 
 Gastric cancer ↑ caveolin-1 strong indicator of poor median overall survival, tumor grade, lymph node involvement, and decreased relapse-free survival in resected gastric cancer Caveolin-1 Poor (135, 136) 
 NSCLC ↑ caveolin-1 expression correlated with pathologic stage, chemoresistance, and decreased overall and disease-free survival Caveolin-1 Poor (138, 139) 
 Prostate cancer ↑ stromal caveolin-1 associated with favorable prognosis, longer survival while expression decreased in malignant tissues and higher tumor stages Caveolin-1 (stromal) Favorable (134) 
 Thyroid cancer ↑ stromal caveolin-1, caveolin-2, and caveolin-3 in anaplastic carcinoma (poor prognosis) compared with papillary thyroid carcinoma and diffuse sclerosing variant of papillary carcinoma (more favorable prognosis) Caveolin-1, -2, and -3 (stromal) Poor (140) 
Protein familyCancer typeSummaryProteinPrognosisReferences
Flotillin Breast cancer ↑ flotillin-2 is significantly correlated with clinical stage, metastasis, and shorter overall survival Flotillin-2 Poor (119) 
 Cervical cancer ↑ flotillin-1 and flotillin-2 corelates with pelvic lymph node metastasis, clinical stage, tumor differentiation, and poor overall survival Flotillin-1 and -2 Poor (120, 121) 
 Colorectal cancer ↑ flotillin-1 and flotillin-2 are associated with tumor volume, depth of invasion, lymph node metastasis, distant metastasis, increased proliferation, and poor survival Flotillin-1 and -2 Poor (125, 126) 
 Liver (hepatocellular and intrahepatic cholangio carcinoma) ↑ flotillin-1 and flotillin-2 in cancerous tissue and positively correlated with tumor size, clinical stage, vascular invasion, lymph node metastasis, and relapse. Flotillin-1 and -2 Poor (123, 124) 
 Nasopharyngeal carcinoma ↑ flotillin-2 promotes tumor progression and is positively associated with a metastatic phenotype and sorter overall survival Flotillin-2 Poor (128, 129) 
 Neuroblastoma ↓ flotillin-1 correlated with poor prognosis and mRNA expression inversely correlate with clinical malignancy grade Flotillin-1 Favorable (130) 
 Small-cell lung cancer ↑ flotillin-1 is highly expressed in small-cell lung cancer and strongly correlates with clinical stage, distant metastasis, and poor survival Flotillin-1 Poor (127) 
 Various ↑ flotillin-1 and flotillin-2 are associated with shorter overall survival, decreased disease-free survival, and increased lymph node metastasis Flotillin-1 and -2 Poor (117, 118) 
 Vulvar squamous cell carcinoma ↑ flotillin-1 predicts poor overall and progressive free survival and is an oncogenic facilitator of inguinal/femoral lymph node metastasis Flotillin-1 Poor (122) 
Caveolin Bladder cancer ↑ caveolin-1 in cancerous tissues and high-grade tumors Caveolin-1 Poor (132, 133) 
 Breast cancer ↑ stromal caveolin-1 in CAFs correlated with increased 5-year survival, low histologic grade while absence or ↓ stromal caveolin-1 increased risk of recurrence and predicted lymph node metastasis Caveolin-1 (stromal and epithelial) Favorable (stromal) (142–145) 
  ↑ epithelial caveolin-1 correlates with high histologic grade, lack of hormone receptors, and decreased survival  Poor (tumoral)  
  Tumor + stromal cancer subtypes had poor survival    
 Breast cancer (brain metastases) ↑ caveolin-1 negatively regulates Stat3, inhibiting brain metastasis from breast cancer Caveolin-1 Favorable (146) 
 Colorectal cancer (liver metastases) ↓ stromal caveolin-1 is associated with decreased disease-free and overall survival after hepatectomy Caveolin-1 (stromal) Favorable (137) 
 Gastric cancer ↑ caveolin-1 strong indicator of poor median overall survival, tumor grade, lymph node involvement, and decreased relapse-free survival in resected gastric cancer Caveolin-1 Poor (135, 136) 
 NSCLC ↑ caveolin-1 expression correlated with pathologic stage, chemoresistance, and decreased overall and disease-free survival Caveolin-1 Poor (138, 139) 
 Prostate cancer ↑ stromal caveolin-1 associated with favorable prognosis, longer survival while expression decreased in malignant tissues and higher tumor stages Caveolin-1 (stromal) Favorable (134) 
 Thyroid cancer ↑ stromal caveolin-1, caveolin-2, and caveolin-3 in anaplastic carcinoma (poor prognosis) compared with papillary thyroid carcinoma and diffuse sclerosing variant of papillary carcinoma (more favorable prognosis) Caveolin-1, -2, and -3 (stromal) Poor (140) 

Flotillins

Flotillin-1 and flotillin-2 are widely believed to be metastatic drivers in a variety of tumors (116). Two recent meta-analyses found that flotillin overexpression predicts poor overall survival, lymph node metastasis, and distant metastasis in a multitude of solid tumors (117, 118). In breast cancer, flotillin-2 mRNA and protein overexpression were indicative of poor prognosis in both early- and late-stage disease (119). Meanwhile, increased flotillin-1/flotillin-2 expression correlated with poor survival and enhanced pelvic, inguinal, and femoral lymph node metastasis in both cervical and vulvar squamous cell carcinoma (120–122). High flotillin-1 and flotillin-2 levels were indicators of aggressive characteristics and poor prognosis of hepatocellular carcinoma and intrahepatic cholangiocarcinoma, respectively (123, 124). In both left and right colorectal cancer, flotillin-1 was overexpressed in cancerous tissue and was associated with tumor volume, differentiation, and proliferation, while flotillin-2 levels were associated with lymph node and distant metastasis (125, 126). Moreover, flotillin-1 and flotillin-2 were found to promote metastasis via TGFβ-mediated EMT induction in patients with small-cell lung cancer and nasopharyngeal carcinoma, respectively (127–129).

While mounting evidence implicates flotillin overexpression with tumor progression and metastasis, one study revealed that low expression of flotillin-1 correlates with poor prognosis in patients with neuroblastoma (130). Flotillin-1 regulated neuroblastoma progression by facilitating binding, endocytosis, and degradation of membrane-localizing anaplastic lymphoma kinase (ALK). Thus, it is possible that this conflicting, antimetastatic role of flotillin-1 may be constrained to subsets of cancers that possess high levels of oncogenic ALK mutants, for example, NSCLC and neuroblastoma (131).

Caveolins

While the oncogenic roles of flotillin-1 and -2 in the clinic are generally agreed upon, there remains contradictory evidence for the clinical role of caveolins, particularly caveolin-1. For example, multiple studies in bladder cancer have demonstrated high caveolin-1 expression is associated with cancer progression, high-grade tumors, and poor patient prognosis (132, 133). Conversely, high stromal caveolin-1 in early prostate cancer was found to correlate with decreased malignancy, longer survival, and favorable prognosis when managed by watchful waiting (134). In gastric cancers, after tumor resection, high caveolin-1 expression correlated with tumor relapse and lymph node metastasis (135, 136). However, in colorectal cancer–derived liver metastasis after hepatectomy, weak stromal caveolin-1 expression was a predictor of poor prognosis (137). Furthermore, enhanced tumoral caveolin-1 correlated with gemcitabine drug resistance and advanced pathologic stage and metastasis in patients with NSCLC (138, 139). Taken together, these studies indicate tumoral caveolin-1 is an indicator of poor prognosis, while stromal caveolin-1 is favorable. While this is true for many studies, in thyroid cancer, expression of stromal caveolin was found to be upregulated in more aggressive carcinoma subtypes (140).

Breast cancer may be the most confounding cancer type when it comes to the clinical relevance of caveolin-1 (141). Overexpression of caveolin-1 in CAFs correlated with increased low histologic grade and favorable prognosis, while absent or depleted stromal caveolin-1 increased the risk of recurrence and predicted lymph node metastasis in early ductal carcinoma in situ (DCIS; refs. 142, 143). In addition, high levels of epithelial caveolin-1 were correlated with more aggressive, TNBC subtypes (144). These findings are supported by a study that examined subgroups of patients with breast cancer with high tumoral caveolin-1 (T++) and weak stromal expression (S−). T(++)/S(−) subgroups showed exceptionally poor clinical outcomes compared with T(++) and S(−) groups taken individually (145). This is consistent with the differential roles of epithelial and stromal caveolin-1 in cancer prognosis discussed above. However, this may not always be the case. One study of metastatic breast cancer found caveolin-1 expression was decreased in brain metastases compared with primary DCIS (146). Furthermore, another study demonstrated that depletion of caveolin-1 in lipid rafts of breast tumors promoted cellular autophagy–mediated cell survival under starvation conditions (147). This supports the multifaceted functionality of caveolins within breast cancer metastasis, warranting further investigation before implementation as a clinical biomarker.

Within the past few decades, numerous studies have implicated lipid rafts as drivers of oncogenic and prometastatic processes. However, the role of rafts in specific areas of metastasis has remained elusive. For example, little is known about how lipid rafts affect CTC survival in the vasculature. While we have presented studies that demonstrate that rafts are vital for anoikis resistance, endothelial cell rolling, and vasculature-related adhesion mechanisms, evidence of this in spontaneous CTCs in animal models remains nonexistent. This is likely a consequence of the aforementioned inadequacies of in vivo and real-time lipid raft detection methods. Moreover, no known study has examined the presence or effect of lipid rafts in primary CTCs isolated from human patients. Given the heterogeneous landscape of CTCs, elucidating the undiscovered roles of rafts and raft-associated proteins may be instrumental in predicting CTC subpopulations that will survive in transit and proceed to colonize secondary tumors (148).

Studies have shown that excessive cholesterol in cancer cells is a biomarker of chemoresistance and stemness, but the role of raft microdomains in CSCs remains largely unknown (149). CD44 remains a prominent CSC marker in a variety of cancers, and we have discussed a multitude of studies demonstrating the propensity of CD44 to coalesce into raft fragments. However, other prominent CSC surface markers, such as CD133, remain understudied despite recent methodologies for isolating CD133+ raft fractions (150). It was recently demonstrated that CD133 localizes into lipid rafts in pancreatic cells to enhance chemoresistance, further motivating investigation into other CSC-related proteins (54). Lipid rafts may be a vital missing link needed to fully understand the formation of CSC phenotypes, while also shedding light into mechanisms of senesce and therapeutic resistance. Furthermore, abrogating the juxtaposition of lipid rafts and stemness-related proteins via raft antagonization may provide a means of reverting CSCs into more druggable cellular phenotypes.

While there has been substantial research done on the contribution of lipid rafts in apoptotic and anoikis pathways, the role of lipid rafts in necroptotic pathways has not been studied sufficiently. This review has discussed the importance of lipid rafts in the MLKL-mediated necroptotic pathway, in response to TNF superfamily ligands. However, it is important to note that necroptosis can also be induced by double-stranded RNA and lipopolysaccharides, through TLR 3 and 4 (151, 152). Necroptosis can also be initiated through IFNs (153). It is currently unknown how and whether lipid rafts play a role in this pathway, perhaps in an MLKL-independent fashion. Leveraging known mechanisms of raft-associated apoptosis may be key for therapeutic exploitation. For example, it was mentioned that many chemotherapeutic agents have been shown to translocate death receptors into lipid rafts to enhance apoptotic signaling upon ligand activation. Clinical evaluation of combination treatments of chemotherapeutics and death-inducing ligands (TRAIL and Fas) may prove effective to overcome mechanisms of therapeutic resistance.

Moving forward, a major challenge will be developing targeted therapeutics and drug delivery strategies to selectively disrupt rafts that facilitate oncogenic and prometastatic processes. While FDA-approved cholesterol lowering drugs exist, they tend to be nonspecific and are not yet approved for cancer therapy (154). In addition, the off-target and downstream signaling implications following statin-mediated cholesterol depletion in cancer have yet to be fully elucidated. Given these translational challenges, we expect not only forthcoming research on raft antagonizing compounds, but raft-targeted delivery modalities as well. For example, one group designed mβCD–hyaluronic acid–ceramide nanoassemblies that selectively target and disrupt CD44-positive lipid rafts (155). Engineering novel drug delivery modalities for raft targeting and regulation will be critical for the development of antimetastatic cancer therapies.

No disclosures were reported.

1.
Simons
K
,
Ikonen
E
. 
Functional rafts in cell membranes
.
Nature
1997
;
387
:
569
72
.
2.
Singer
SJ
,
Nicolson
GL
. 
The fluid mosaic model of the structure of cell membranes
.
Science
1972
;
175
:
720
31
.
3.
Sezgin
E
,
Levental
I
,
Mayor
S
,
Eggeling
C
. 
The mystery of membrane organization: composition, regulation and roles of lipid rafts
.
Nat Rev Mol Cell Biol
2017
;
18
:
361
74
.
4.
Simons
K
,
Ehehalt
R
. 
Cholesterol, lipid rafts, and disease
.
J Clin Invest
2002
;
110
:
597
603
.
5.
Hanzal-Bayer
MF
,
Hancock
JF
. 
Lipid rafts and membrane traffic
.
FEBS Lett
2007
;
581
:
2098
104
.
6.
Karnovsky
MJ
,
Kleinfeld
AM
,
Hoover
RL
,
Klausner
RD
. 
The concept of lipid domains in membranes
.
J Cell Biol
1982
;
94
:
1
6
.
7.
Chamberlain
LH
. 
Detergents as tools for the purification and classification of lipid rafts
.
FEBS Lett
2004
;
559
:
1
5
.
8.
Brown
DA
,
London
E
. 
Functions of lipid rafts in biological membranes
.
Annu Rev Cell Dev Biol
1998
;
14
:
111
36
.
9.
Klymchenko
AS
,
Kreder
R
. 
Fluorescent probes for lipid rafts: from model membranes to living cells
.
Chem Biol
2014
;
21
:
97
113
.
10.
Martinez-Outschoorn
UE
,
Sotgia
F
,
Lisanti
MP
. 
Caveolae and signalling in cancer
.
Nat Rev Cancer
2015
;
15
:
225
37
.
11.
Galbiati
F
,
Razani
B
,
Lisanti
MP
. 
Emerging themes in lipid rafts and caveolae
.
Cell
2001
;
106
:
403
11
.
12.
de Laurentiis
A
,
Donovan
L
,
Arcaro
A
. 
Lipid rafts and caveolae in signaling by growth factor receptors
.
Open Biochem J
2007
;
1
:
12
32
.
13.
Otto
GP
,
Nichols
BJ
. 
The roles of flotillin microdomains–endocytosis and beyond
.
J Cell Sci
2011
;
124
:
3933
40
.
14.
Simons
K
,
Toomre
D
. 
Lipid rafts and signal transduction
.
Nat Rev Mol Cell Biol
2000
;
1
:
31
9
.
15.
Shi
D
,
Lv
X
,
Zhang
Z
,
Yang
X
,
Zhou
Z
,
Zhang
L
, et al
Smoothened oligomerization/higher order clustering in lipid rafts is essential for high Hedgehog activity transduction
.
J Biol Chem
2013
;
288
:
12605
14
.
16.
Staubach
S
,
Hanisch
F-G
. 
Lipid rafts: signaling and sorting platforms of cells and their roles in cancer
.
Expert Rev Proteomics
2011
;
8
:
263
77
.
17.
Valastyan
S
,
Weinberg
RA
. 
Tumor metastasis: molecular insights and evolving paradigms
.
Cell
2011
;
147
:
275
92
.
18.
Mehlen
P
,
Puisieux
A
. 
Metastasis: a question of life or death
.
Nat Rev Cancer
2006
;
6
:
449
58
.
19.
Vincent
L
,
Chen
W
,
Hong
L
,
Mirshahi
F
,
Mishal
Z
,
Mirshahi-Khorassani
T
, et al
Inhibition of endothelial cell migration by cerivastatin, an HMG-CoA reductase inhibitor: contribution to its anti-angiogenic effect
.
FEBS Lett
2001
;
495
:
159
66
.
20.
Badana
A
,
Chintala
M
,
Varikuti
G
,
Pudi
N
,
Kumari
S
,
Kappala
VR
, et al
Lipid raft integrity is required for survival of triple negative breast cancer cells
.
J Breast Cancer
2016
;
19
:
372
84
.
21.
Raghu
H
,
Sodadasu
PK
,
Malla
RR
,
Gondi
CS
,
Estes
N
,
Rao
JS
. 
Localization of uPAR and MMP-9 in lipid rafts is critical for migration, invasion and angiogenesis in human breast cancer cells
.
BMC Cancer
2010
;
10
:
647
.
22.
Ben-Zaken
O
,
Gingis-Velitski
S
,
Vlodavsky
I
,
Ilan
N
. 
Heparanase induces Akt phosphorylation via a lipid raft receptor
.
Biochem Biophys Res Commun
2007
;
361
:
829
34
.
23.
Chatterjee
S
,
Heukamp
LC
,
Siobal
M
,
Schöttle
J
,
Wieczorek
C
,
Peifer
M
, et al
Tumor VEGF:VEGFR2 autocrine feed-forward loop triggers angiogenesis in lung cancer
.
J Clin Invest
2013
;
123
:
1732
40
.
24.
Noghero
A
,
Perino
A
,
Seano
G
,
Saglio
E
,
Lo Sasso
G
,
Veglio
F
, et al
Liver X receptor activation reduces angiogenesis by impairing lipid raft localization and signaling of vascular endothelial growth factor receptor-2
.
Arterioscler Thromb Vasc Biol
2012
;
32
:
2280
8
.
25.
Tahir
SA
,
Park
S
,
Thompson
TC
. 
Caveolin-1 regulates VEGF-stimulated angiogenic activities in prostate cancer and endothelial cells
.
Cancer Biol Ther
2009
;
8
:
2286
96
.
26.
Nomura
S
,
Iwata
S
,
Hatano
R
,
Komiya
E
,
Dang
NH
,
Iwao
N
, et al
Inhibition of VEGF-dependent angiogenesis by the anti-CD82 monoclonal antibody 4F9 through regulation of lipid raft microdomains
.
Biochem Biophys Res Commun
2016
;
474
:
111
7
.
27.
Wang
X
,
Zhang
Y
,
Zhao
Y
,
Liang
Y
,
Xiang
C
,
Zhou
H
, et al
CD24 promoted cancer cell angiogenesis via Hsp90-mediated STAT3/VEGF signaling pathway in colorectal cancer
.
Oncotarget
2016
;
7
:
55663
76
.
28.
Hoshino
A
,
Costa-Silva
B
,
Shen
T-L
,
Rodrigues
G
,
Hashimoto
A
,
Tesic Mark
M
, et al
Tumour exosome integrins determine organotropic metastasis
.
Nature
2015
;
527
:
329
35
.
29.
He
L
,
Zhu
W
,
Chen
Q
,
Yuan
Y
,
Wang
Y
,
Wang
J
, et al
Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis
.
Theranostics
2019
;
9
:
8206
20
.
30.
Jin
H
,
He
Y
,
Zhao
P
,
Hu
Y
,
Tao
J
,
Chen
J
, et al
Targeting lipid metabolism to overcome EMT-associated drug resistance via integrin β3/FAK pathway and tumor-associated macrophage repolarization using legumain-activatable delivery
.
Theranostics
2019
;
9
:
265
78
.
31.
Cheng
Y
,
Song
Y
,
Qu
J
,
Che
X
,
Song
N
,
Fan
Y
, et al
The chemokine receptor CXCR4 and c-MET cooperatively promote epithelial-mesenchymal transition in gastric cancer cells
.
Transl Oncol
2018
;
11
:
487
97
.
32.
Zuo
W
,
Chen
Y-G
. 
Specific activation of mitogen-activated protein kinase by transforming growth factor-β receptors in lipid rafts is required for epithelial cell plasticity
.
Mol Biol Cell
2009
;
20
:
1020
9
.
33.
Xu
J
,
Lamouille
S
,
Derynck
R
. 
TGF-β-induced epithelial to mesenchymal transition
.
Cell Res
2009
;
19
:
156
72
.
34.
Hung
C-M
,
Kuo
D-H
,
Chou
C-H
,
Su
Y-C
,
Ho
C-T
,
Way
T-D
. 
Osthole suppresses hepatocyte growth factor (HGF)-induced epithelial-mesenchymal transition via repression of the c-Met/Akt/mTOR pathway in human breast cancer cells
.
J Agric Food Chem
2011
;
59
:
9683
90
.
35.
Fernández-Muñoz
B
,
Yurrita
MM
,
Martín-Villar
E
,
Carrasco-Ramírez
P
,
Megías
D
,
Renart
J
, et al
The transmembrane domain of podoplanin is required for its association with lipid rafts and the induction of epithelial-mesenchymal transition
.
Int J Biochem Cell Biol
2011
;
43
:
886
96
.
36.
Liang
W
,
Hao
Z
,
Han
J-L
,
Zhu
D-J
,
Jin
Z-F
,
Xie
W-L
. 
CAV-1 contributes to bladder cancer progression by inducing epithelial-to-mesenchymal transition
.
Urol Oncol Semin Orig Investig
2014
;
32
:
855
63
.
37.
Li
Q
,
Peng
J
,
Li
X
,
Leng
A
,
Liu
T
. 
miR-449a targets Flot2 and inhibits gastric cancer invasion by inhibiting TGF-β-mediated EMT
.
Diagn Pathol
2015
;
10
:
202
.
38.
Tisza
MJ
,
Zhao
W
,
Fuentes
JSR
,
Prijic
S
,
Chen
X
,
Levental
I
, et al
Motility and stem cell properties induced by the epithelial-mesenchymal transition require destabilization of lipid rafts
.
Oncotarget
2016
;
7
:
51553
68
.
39.
Head
BP
,
Patel
HH
,
Insel
PA
. 
Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling
.
Biochim Biophys Acta
2014
;
1838
:
532
45
.
40.
Wang
R
,
Bi
J
,
Ampah
KK
,
Ba
X
,
Liu
W
,
Zeng
X
. 
Lipid rafts control human melanoma cell migration by regulating focal adhesion disassembly
.
Biochim Biophys Acta
2013
;
1833
:
3195
205
.
41.
Li
G
,
Sasaki
T
,
Asahina
S
,
Roy
MC
,
Mochizuki
T
,
Koizumi
K
, et al
Patching of lipid rafts by molecular self-assembled nanofibrils suppresses cancer cell migration
.
Chem
2017
;
2
:
283
98
.
42.
Chantôme
A
,
Potier-Cartereau
M
,
Clarysse
L
,
Fromont
G
,
Marionneau-Lambot
S
,
Guéguinou
M
, et al
Pivotal role of the lipid raft SK3–orai1 complex in human cancer cell migration and bone metastases
.
Cancer Res
2013
;
73
:
4852
61
.
43.
Gialeli
C
,
Theocharis
AD
,
Karamanos
NK
. 
Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting
.
FEBS J
2011
;
278
:
16
27
.
44.
Zhang
Q
,
Furukawa
K
,
Chen
H-H
,
Sakakibara
T
,
Urano
T
,
Furukawa
K
. 
Metastatic potential of mouse Lewis lung cancer cells is regulated via ganglioside GM1 by modulating the matrix metalloprotease-9 localization in lipid rafts
.
J Biol Chem
2006
;
281
:
18145
55
.
45.
Yang
Y-F
,
Jan
Y-H
,
Liu
Y-P
,
Yang
C-J
,
Su
C-Y
,
Chang
Y-C
, et al
Squalene synthase induces tumor necrosis factor receptor 1 enrichment in lipid rafts to promote lung cancer metastasis
.
Am J Respir Crit Care Med
2014
;
190
:
675
87
.
46.
Brand
S
,
Dambacher
J
,
Beigel
F
,
Olszak
T
,
Diebold
J
,
Otte
J-M
, et al
CXCR4 and CXCL12 are inversely expressed in colorectal cancer cells and modulate cancer cell migration, invasion and MMP-9 activation
.
Exp Cell Res
2005
;
310
:
117
30
.
47.
Arya
M
,
Ahmed
H
,
Silhi
N
,
Williamson
M
,
Patel
HRH
. 
Clinical importance and therapeutic implications of the pivotal CXCL12-CXCR4 (chemokine ligand-receptor) interaction in cancer cell migration
.
Tumor Biol
2007
;
28
:
123
31
.
48.
Conley-LaComb
MK
,
Semaan
L
,
Singareddy
R
,
Li
Y
,
Heath
EI
,
Kim
S
, et al
Pharmacological targeting of CXCL12/CXCR4 signaling in prostate cancer bone metastasis
.
Mol Cancer
2016
;
15
:
68
.
49.
Sbrissa
D
,
Semaan
L
,
Govindarajan
B
,
Li
Y
,
Caruthers
NJ
,
Stemmer
PM
, et al
A novel cross-talk between CXCR4 and PI4KIIIα in prostate cancer cells
.
Oncogene
2019
;
38
:
332
44
.
50.
Liu
Y
,
Sun
R
,
Wan
W
,
Wang
J
,
Oppenheim
JJ
,
Chen
L
, et al
The involvement of lipid rafts in epidermal growth factor-induced chemotaxis of breast cancer cells
.
Mol Membr Biol
2007
;
24
:
91
101
.
51.
Grolez
GP
,
Gordiendko
DV
,
Clarisse
M
,
Hammadi
M
,
Desruelles
E
,
Fromont
G
, et al
TRPM8-androgen receptor association within lipid rafts promotes prostate cancer cell migration
.
Cell Death Dis
2019
;
10
:
1
17
.
52.
Xie
G
,
Li
J
,
Chen
J
,
Tang
X
,
Wu
S
,
Liao
C
. 
Knockdown of flotillin-2 impairs the proliferation of breast cancer cells through modulation of Akt/FOXO signaling
.
Oncol Rep
2015
;
33
:
2285
90
.
53.
Berger
T
,
Ueda
T
,
Arpaia
E
,
Chio
IIC
,
Shirdel
EA
,
Jurisica
I
, et al
Flotillin-2 deficiency leads to reduced lung metastases in a mouse breast cancer model
.
Oncogene
2013
;
32
:
4989
94
.
54.
Gupta
VK
,
Sharma
NS
,
Kesh
K
,
Dauer
P
,
Nomura
A
,
Giri
B
, et al
Metastasis and chemoresistance in CD133 expressing pancreatic cancer cells are dependent on their lipid raft integrity
.
Cancer Lett
2018
;
439
:
101
12
.
55.
Reymond
N
,
d'Água
BB
,
Ridley
AJ
. 
Crossing the endothelial barrier during metastasis
.
Nat Rev Cancer
2013
;
13
:
858
70
.
56.
Roland
CL
,
Harken
AH
,
Sarr
MG
,
Barnett
CC
. 
ICAM-1 expression determines malignant potential of cancer
.
Surgery
2007
;
141
:
705
7
.
57.
Rahn
JJ
,
Chow
JW
,
Horne
GJ
,
Mah
BK
,
Emerman
JT
,
Hoffman
P
, et al
MUC1 mediates transendothelial migration in vitro by ligating endothelial cell ICAM-1
.
Clin Exp Metastasis
2005
;
22
:
475
83
.
58.
Shi
H
,
Fang
W
,
Liu
M
,
Fu
D
. 
Complement component 1, q subcomponent binding protein (C1QBP) in lipid rafts mediates hepatic metastasis of pancreatic cancer by regulating IGF-1/IGF-1R signaling
.
Int J Cancer
2017
;
141
:
1389
401
.
59.
Tokui
N
,
Yoneyama
MS
,
Hatakeyama
S
,
Yamamoto
H
,
Koie
T
,
Saitoh
H
, et al
Extravasation during bladder cancer metastasis requires cortactin-mediated invadopodia formation
.
Mol Med Rep
2014
;
9
:
1142
6
.
60.
Williams
KC
,
Cepeda
MA
,
Javed
S
,
Searle
K
,
Parkins
KM
,
Makela
AV
, et al
Invadopodia are chemosensing protrusions that guide cancer cell extravasation to promote brain tropism in metastasis
.
Oncogene
2019
;
38
:
3598
615
.
61.
Gligorijevic
B
,
Wyckoff
J
,
Yamaguchi
H
,
Wang
Y
,
Roussos
ET
,
Condeelis
J
. 
N-WASP-mediated invadopodium formation is involved in intravasation and lung metastasis of mammary tumors
.
J Cell Sci
2012
;
125
:
724
34
.
62.
Leong
HS
,
Robertson
AE
,
Stoletov
K
,
Leith
SJ
,
Chin
CA
,
Chien
AE
, et al
Invadopodia are required for cancer cell extravasation and are a therapeutic target for metastasis
.
Cell Rep
2014
;
8
:
1558
70
.
63.
Knopf
JD
,
Tholen
S
,
Koczorowska
MM
,
De Wever
O
,
Biniossek
ML
,
Schilling
O
. 
The stromal cell-surface protease fibroblast activation protein-α localizes to lipid rafts and is recruited to invadopodia
.
Biochim Biophys Acta
2015
;
1853
:
2515
25
.
64.
Yamaguchi
H
,
Takeo
Y
,
Yoshida
S
,
Kouchi
Z
,
Nakamura
Y
,
Fukami
K
. 
Lipid rafts and caveolin-1 are required for invadopodia formation and extracellular matrix degradation by human breast cancer cells
.
Cancer Res
2009
;
69
:
8594
602
.
65.
Yang
H
,
Guan
L
,
Li
S
,
Jiang
Y
,
Xiong
N
,
Li
L
, et al
Mechanosensitive caveolin-1 activation-induced PI3K/Akt/mTOR signaling pathway promotes breast cancer motility, invadopodia formation and metastasis in vivo
.
Oncotarget
2016
;
7
:
16227
47
.
66.
Martín-Villar
E
,
Borda-d'Agua
B
,
Carrasco-Ramirez
P
,
Renart
J
,
Parsons
M
,
Quintanilla
M
, et al
Podoplanin mediates ECM degradation by squamous carcinoma cells through control of invadopodia stability
.
Oncogene
2015
;
34
:
4531
44
.
67.
Bendas
G
,
Borsig
L
. 
Cancer cell adhesion and metastasis: selectins, integrins, and the inhibitory potential of heparins
.
Int J Cell Biol
2012
;
2012
:
e676731
.
68.
Sun
M
,
Northup
N
,
Marga
F
,
Huber
T
,
Byfield
FJ
,
Levitan
I
, et al
The effect of cellular cholesterol on membrane-cytoskeleton adhesion
.
J Cell Sci
2007
;
120
:
2223
31
.
69.
Eich
C
,
Manzo
C
,
de Keijzer
S
,
Bakker
G-J
,
Reinieren-Beeren
I
,
García-Parajo
MF
, et al
Changes in membrane sphingolipid composition modulate dynamics and adhesion of integrin nanoclusters
.
Sci Rep
2016
;
6
:
20693
.
70.
Gopalakrishna
P
,
Rangaraj
N
,
Pande
G
. 
Cholesterol alters the interaction of glycosphingolipid GM3 with alpha5beta1 integrin and increases integrin-mediated cell adhesion to fibronectin
.
Exp Cell Res
2004
;
300
:
43
53
.
71.
Yates
AJ
,
Rampersaud
A
. 
Sphingolipids as receptor modulators: an overview
.
Ann N Y Acad Sci
1998
;
845
:
57
71
.
72.
Okada
Y
,
Nishikawa
J
,
Semma
M
,
Ichikawa
A
. 
Role of lipid raft components and actin cytoskeleton in fibronectin-binding, surface expression, and de novo synthesis of integrin subunits in PGE2- or 8-Br-cAMP-stimulated mastocytoma P-815 cells
.
Biochem Pharmacol
2014
;
88
:
364
71
.
73.
Leitinger
B
,
Hogg
N
. 
The involvement of lipid rafts in the regulation of integrin function
.
J Cell Sci
2002
;
115
:
963
72
.
74.
Marhaba
R
,
Zöller
M
. 
CD44 in cancer progression: adhesion, migration and growth regulation
.
J Mol Histol
2004
;
35
:
211
31
.
75.
Draffin
JE
,
McFarlane
S
,
Hill
A
,
Johnston
PG
,
Waugh
DJJ
. 
CD44 potentiates the adherence of metastatic prostate and breast cancer cells to bone marrow endothelial cells
.
Cancer Res
2004
;
64
:
5702
11
.
76.
Herrera-Gayol
A
,
Jothy
S
. 
CD44 modulates Hs578T human breast cancer cell adhesion, migration, and invasiveness
.
Exp Mol Pathol
1999
;
66
:
99
108
.
77.
Wang
H
,
Zhu
Y
,
Zhao
M
,
Wu
C
,
Zhang
P
,
Tang
L
, et al
miRNA-29c suppresses lung cancer cell adhesion to extracellular matrix and metastasis by targeting integrin β1 and matrix metalloproteinase2 (MMP2)
.
PLoS One
2013
;
8
:
e70192
.
78.
Albelda
SM
. 
Role of integrins and other cell adhesion molecules in tumor progression and metastasis
.
Lab Invest
1993
;
68
:
4
17
.
79.
Felding-Habermann
B
,
O'Toole
TE
,
Smith
JW
,
Fransvea
E
,
Ruggeri
ZM
,
Ginsberg
MH
, et al
Integrin activation controls metastasis in human breast cancer
.
Proc Natl Acad Sci U S A
2001
;
98
:
1853
8
.
80.
Murai
T
,
Maruyama
Y
,
Mio
K
,
Nishiyama
H
,
Suga
M
,
Sato
C
. 
Low cholesterol triggers membrane microdomain-dependent CD44 shedding and suppresses tumor cell migration
.
J Biol Chem
2011
;
286
:
1999
2007
.
81.
Mohammadalipour
A
,
Showalter
C
,
Muturi
HT
,
Sharma
V
,
Farnoud
AM
,
Puri
V
, et al
Abstract 98: Cell membrane cholesterol modulates lung cancer cell adhesion and rolling on E-selectin
.
Cancer Res
2018
;
78
:
98
.
82.
Lee
J-L
,
Wang
M-J
,
Sudhir
P-R
,
Chen
J-Y
. 
CD44 engagement promotes matrix-derived survival through the CD44-SRC-integrin axis in lipid rafts
.
Mol Cell Biol
2008
;
28
:
5710
23
.
83.
Hitosugi
T
,
Sato
M
,
Sasaki
K
,
Umezawa
Y
. 
Lipid raft specific knockdown of SRC family kinase activity inhibits cell adhesion and cell cycle progression of breast cancer cells
.
Cancer Res
2007
;
67
:
8139
48
.
84.
Baumann
P
,
Thiele
W
,
Cremers
N
,
Muppala
S
,
Krachulec
J
,
Diefenbacher
M
, et al
CD24 interacts with and promotes the activity of c-src within lipid rafts in breast cancer cells, thereby increasing integrin-dependent adhesion
.
Cell Mol Life Sci
2012
;
69
:
435
48
.
85.
Huang
Q
,
Shen
H-M
,
Shui
G
,
Wenk
MR
,
Ong
C-N
. 
Emodin inhibits tumor cell adhesion through disruption of the membrane lipid Raft-associated integrin signaling pathway
.
Cancer Res
2006
;
66
:
5807
15
.
86.
Li
C
,
Lu
N
,
Qi
Q
,
Li
F
,
Ling
Y
,
Chen
Y
, et al
Gambogic acid inhibits tumor cell adhesion by suppressing integrin β1 and membrane lipid rafts-associated integrin signaling pathway
.
Biochem Pharmacol
2011
;
82
:
1873
83
.
87.
Babina
IS
,
McSherry
EA
,
Donatello
S
,
Hill
AD
,
Hopkins
AM
. 
A novel mechanism of regulating breast cancer cell migration via palmitoylation-dependent alterations in the lipid raft affiliation of CD44
.
Breast Cancer Res
2014
;
16
:
R19
.
88.
Sun
F
,
Schroer
CFE
,
Palacios
CR
,
Xu
L
,
Luo
S-Z
,
Marrink
SJ
. 
Molecular mechanism for bidirectional regulation of CD44 for lipid raft affiliation by palmitoylations and PIP2
.
PLOS Comput Biol
2020
;
16
:
e1007777
.
89.
Renehan
AG
,
Booth
C
,
Potten
CS
. 
What is apoptosis, and why is it important?
BMJ
2001
;
322
:
1536
8
.
90.
Borges
HL
,
Linden
R
,
Wang
JY
. 
DNA damage-induced cell death
.
Cell Res
2008
;
18
:
17
26
.
91.
Fernald
K
,
Kurokawa
M
. 
Evading apoptosis in cancer
.
Trends Cell Biol
2013
;
23
:
620
33
.
92.
Syntichaki
P
,
Tavernarakis
N
. 
Death by necrosis
.
EMBO Rep
2002
;
3
:
604
9
.
93.
Proskuryakov
SYa
,
Gabai
VL
,
Konoplyannikov
AG
. 
Necrosis is an active and controlled form of programmed cell death
.
Biochem Mosc
2002
;
67
:
387
408
.
94.
Weber
K
,
Roelandt
R
,
Bruggeman
I
,
Estornes
Y
,
Vandenabeele
P
. 
Nuclear RIPK3 and MLKL contribute to cytosolic necrosome formation and necroptosis
.
Commun Biol
2018
;
1
:
1
13
.
95.
Chen
X
,
Li
W
,
Ren
J
,
Huang
D
,
He
W
,
Song
Y
, et al
Translocation of mixed lineage kinase domain-like protein to plasma membrane leads to necrotic cell death
.
Cell Res
2014
;
24
:
105
21
.
96.
Dondelinger
Y
,
Declercq
W
,
Montessuit
S
,
Roelandt
R
,
Goncalves
A
,
Bruggeman
I
, et al
MLKL compromises plasma membrane integrity by binding to phosphatidylinositol phosphates
.
Cell Rep
2014
;
7
:
971
81
.
97.
Zhang
X
,
Kitatani
K
,
Toyoshima
M
,
Ishibashi
M
,
Usui
T
,
Minato
J
, et al
Ceramide nanoliposomes as a MLKL-dependent, necroptosis-inducing, chemotherapeutic reagent in ovarian cancer
.
Mol Cancer Ther
2018
;
17
:
50
9
.
98.
Elmore
S
. 
Apoptosis: a review of programmed cell death
.
Toxicol Pathol
2007
;
35
:
495
516
.
99.
Larsen
BD
,
Sørensen
CS
. 
The caspase-activated DNase: apoptosis and beyond
.
FEBS J
2017
;
284
:
1160
70
.
100.
Gajate
C
,
Mollinedo
F
. 
The antitumor ether lipid ET-18-OCH(3) induces apoptosis through translocation and capping of Fas/CD95 into membrane rafts in human leukemic cells
.
Blood
2001
;
98
:
3860
3
.
101.
Reis-Sobreiro
M
,
Gajate
C
,
Mollinedo
F
. 
Involvement of mitochondria and recruitment of Fas/CD95 signaling in lipid rafts in resveratrol-mediated antimyeloma and antileukemia actions
.
Oncogene
2009
;
28
:
3221
34
.
102.
Xiao
W
,
Ishdorj
G
,
Sun
J
,
Johnston
JB
,
Gibson
SB
. 
Death receptor 4 is preferentially recruited to lipid rafts in chronic lymphocytic leukemia cells contributing to tumor necrosis related apoptosis inducing ligand-induced synergistic apoptotic responses
.
Leuk Lymphoma
2011
;
52
:
1290
301
.
103.
Marconi
M
,
Ascione
B
,
Ciarlo
L
,
Vona
R
,
Garofalo
T
,
Sorice
M
, et al
Constitutive localization of DR4 in lipid rafts is mandatory for TRAIL-induced apoptosis in B-cell hematologic malignancies
.
Cell Death Dis
2013
;
4
:
e863
.
104.
Ouyang
W
,
Yang
C
,
Liu
Y
,
Xiong
J
,
Zhang
J
,
Zhong
Y
, et al
Redistribution of DR4 and DR5 in lipid rafts accounts for the sensitivity to TRAIL in NSCLC cells
.
Int J Oncol
2011
;
39
:
1577
86
.
105.
Song
JH
,
Tse
MCL
,
Bellail
A
,
Phuphanich
S
,
Khuri
F
,
Kneteman
NM
, et al
Lipid rafts and nonrafts mediate tumor necrosis factor–related apoptosis-inducing ligand–induced apoptotic and nonapoptotic signals in non–small cell lung carcinoma cells
.
Cancer Res
2007
;
67
:
6946
55
.
106.
Delmas
D
,
Rébé
C
,
Micheau
O
,
Athias
A
,
Gambert
P
,
Grazide
S
, et al
Redistribution of CD95, DR4 and DR5 in rafts accounts for the synergistic toxicity of resveratrol and death receptor ligands in colon carcinoma cells
.
Oncogene
2004
;
23
:
8979
86
.
107.
Xu
L
,
Qu
X
,
Zhang
Y
,
Hu
X
,
Yang
X
,
Hou
K
, et al
Oxaliplatin enhances TRAIL-induced apoptosis in gastric cancer cells by CBL-regulated death receptor redistribution in lipid rafts
.
FEBS Lett
2009
;
583
:
943
8
.
108.
Mollinedo
F
,
Gajate
C
. 
Lipid rafts as major platforms for signaling regulation in cancer
.
Adv Biol Regul
2015
;
57
:
130
46
.
109.
Mollinedo
F
,
Gajate
C
. 
Lipid rafts, death receptors and CASMERs: new insights for cancer therapy
.
Future Oncol
2010
;
6
:
491
4
.
110.
Gao
X
,
Zhang
J
. 
Spatiotemporal analysis of differential Akt regulation in plasma membrane microdomains
.
Mol Biol Cell
2008
;
19
:
4366
73
.
111.
Kim
Y-N
,
Koo
KH
,
Sung
JY
,
Yun
U-J
,
Kim
H
. 
Anoikis resistance: an essential prerequisite for tumor metastasis
.
Int J Cell Biol
2012
;
2012
:
306879
.
112.
Gilmore
AP
. 
Anoikis
.
Cell Death Differ
2005
;
12
:
1473
7
.
113.
Li
YC
,
Park
MJ
,
Ye
S-K
,
Kim
C-W
,
Kim
Y-N
. 
Elevated levels of cholesterol-rich lipid rafts in cancer cells are correlated with apoptosis sensitivity induced by cholesterol-depleting agents
.
Am J Pathol
2006
;
168
:
1107
18
.
114.
Park
E-K
,
Park
MJ
,
Lee
S-H
,
Li
YC
,
Kim
J
,
Lee
J-S
, et al
Cholesterol depletion induces anoikis-like apoptosis via FAK down-regulation and caveolae internalization
.
J Pathol
2009
;
218
:
337
49
.
115.
Lee
S-H
,
Koo
KH
,
Park
J-W
,
Kim
H-J
,
Ye
S-K
,
Park
JB
, et al
HIF-1 is induced via EGFR activation and mediates resistance to anoikis-like cell death under lipid rafts/caveolae-disrupting stress
.
Carcinogenesis
2009
;
30
:
1997
2004
.
116.
Liu
X
,
Liu
W
,
Wang
L
,
Zhu
B
,
Shi
X
,
Peng
Z
, et al
Roles of flotillins in tumors
.
J Zhejiang Univ Sci B
2018
;
19
:
171
82
.
117.
Deng
Y
,
Ge
P
,
Tian
T
,
Dai
C
,
Wang
M
,
Lin
S
, et al
Prognostic value of flotillins (flotillin-1 and flotillin-2) in human cancers: a meta-analysis
.
Clin Chim Acta Int J Clin Chem
2018
;
481
:
90
8
.
118.
Ou
Y
,
Liu
F
,
Chen
F
,
Zhu
Z
. 
Prognostic value of Flotillin-1 expression in patients with solid tumors
.
Oncotarget
2017
;
8
:
52665
77
.
119.
Wang
X
,
Yang
Q
,
Guo
L
,
Li
X-H
,
Zhao
X-H
,
Song
L-B
, et al
Flotillin-2 is associated with breast cancer progression and poor survival outcomes
.
J Transl Med
2013
;
11
:
190
.
120.
Li
Z
,
Yang
Y
,
Gao
Y
,
Wu
X
,
Yang
X
,
Zhu
Y
, et al
Elevated expression of flotillin-1 is associated with lymph node metastasis and poor prognosis in early-stage cervical cancer
.
Am J Cancer Res
2015
;
6
:
38
50
.
121.
Liu
Y
,
Lin
L
,
Huang
Z
,
Ji
B
,
Mei
S
,
Lin
Y
, et al
High expression of flotillin-2 is associated with poor clinical survival in cervical carcinoma
.
Int J Clin Exp Pathol
2015
;
8
:
622
8
.
122.
Gao
Y
,
Wu
X
,
Yang
Y
,
Ruan
Y
,
Yang
X
,
Zhu
Y
, et al
High expression of flotillin-1 is associated with lymph node metastasis and poor prognosis in vulvar squamous cell carcinoma
.
Int J Clin Exp Pathol
2016
;
9
:
9538
46
.
123.
Zhang
S-H
,
Wang
C-J
,
Shi
L
,
Li
X-H
,
Zhou
J
,
Song
L-B
, et al
High expression of FLOT1 is associated with progression and poor prognosis in hepatocellular carcinoma
.
PLoS One
2013
;
8
:
e64709
.
124.
Xu
Z
,
Wang
T
,
Song
H
,
Jiang
X
. 
Flotillin-2 predicts poor prognosis and promotes tumor invasion in intrahepatic cholangiocarcinoma
.
Oncol Lett
2020
;
19
:
2243
50
.
125.
Baig
N
,
Li
Z
,
Lu
J
,
Chen
H
,
Yu
S
,
Li
T
, et al
Clinical significance and comparison of flotillin 1 expression in left and right colon cancer
.
Oncol Lett
2019
;
18
:
997
1004
.
126.
Li
T
,
Cao
C
,
Xiong
Q
,
Liu
D
. 
FLOT2 overexpression is associated with the progression and prognosis of human colorectal cancer
.
Oncol Lett
2019
;
17
:
2802
8
.
127.
Zhao
L
,
Li
J
,
Liu
Y
,
Zhou
W
,
Shan
Y
,
Fan
X
, et al
Flotillin1 promotes EMT of human small cell lung cancer via TGF-β signaling pathway
.
Cancer Biol Med
2018
;
15
:
400
14
.
128.
Liu
J
,
Huang
W
,
Ren
C
,
Wen
Q
,
Liu
W
,
Yang
X
, et al
Flotillin-2 promotes metastasis of nasopharyngeal carcinoma by activating NF-κB and PI3K/Akt3 signaling pathways
.
Sci Rep
2015
;
5
:
11614
.
129.
Zhao
L
,
Shi
M
,
Duan
J
,
Ma
H
,
Wu
Z
,
Chen
X
, et al
Flotillin-2 role in nasopharyngeal carcinoma metastasis and correlation with poor survival outcomes
.
J Clin Oncol
2014
;
32
:
e17050
.
130.
Tomiyama
A
,
Uekita
T
,
Kamata
R
,
Sasaki
K
,
Takita
J
,
Ohira
M
, et al
Flotillin-1 regulates oncogenic signaling in neuroblastoma cells by regulating ALK membrane association
.
Cancer Res
2014
;
74
:
3790
801
.
131.
Umapathy
G
,
Mendoza‐Garcia
P
,
Hallberg
B
,
Palmer
RH
. 
Targeting anaplastic lymphoma kinase in neuroblastoma
.
APMIS
2019
;
127
:
288
302
.
132.
Raja
SA
,
Shah
STA
,
Tariq
A
,
Bibi
N
,
Sughra
K
,
Yousuf
A
, et al
Caveolin-1 and dynamin-2 overexpression is associated with the progression of bladder cancer
.
Oncol Lett
2019
;
18
:
219
26
.
133.
Rajjayabun
PH
,
Garg
S
,
Durkan
GC
,
Charlton
R
,
Robinson
MC
,
Mellon
JK
. 
Caveolin-1 expression is associated with high-grade bladder cancer
.
Urology
2001
;
58
:
811
4
.
134.
Hammarsten
P
,
Dahl Scherdin
T
,
Hägglöf
C
,
Andersson
P
,
Wikström
P
,
Stattin
P
, et al
High caveolin-1 expression in tumor stroma is associated with a favourable outcome in prostate cancer patients managed by watchful waiting
.
PLoS One
2016
;
11
:
e0164016
.
135.
Sun
DS
,
Hong
SA
,
Won
HS
,
Yoo
SH
,
Lee
HH
,
Kim
O
, et al
Prognostic value of metastatic tumoral caveolin-1 expression in patients with resected gastric cancer
.
Gastroenterol Res Pract
2017
;
2017
:
e5905173
.
136.
Seker
M
,
Aydin
D
,
Bilici
A
,
Yavuzer
D
,
Ozgun
MG
,
Ozcelik
M
, et al
Correlation of caveolin-1 expression with prognosis in patients with gastric cancer after gastrectomy
.
Oncol Res Treat
2017
;
40
:
185
90
.
137.
Neofytou
K
,
Pikoulis
E
,
Petrou
A
,
Agrogiannis
G
,
Petrides
C
,
Papakonstandinou
I
, et al
Weak stromal caveolin-1 expression in colorectal liver metastases predicts poor prognosis after hepatectomy for liver-only colorectal metastases
.
Sci Rep
2017
;
7
:
2058
.
138.
Yoo
S-H
,
Park
YS
,
Kim
H-R
,
Sung
SW
,
Kim
JH
,
Shim
YS
, et al
Expression of caveolin-1 is associated with poor prognosis of patients with squamous cell carcinoma of the lung
.
Lung Cancer Amst Neth
2003
;
42
:
195
202
.
139.
Ho
C-C
,
Kuo
S-H
,
Huang
P-H
,
Huang
H-Y
,
Yang
C-H
,
Yang
P-C
. 
Caveolin-1 expression is significantly associated with drug resistance and poor prognosis in advanced non-small cell lung cancer patients treated with gemcitabine-based chemotherapy
.
Lung Cancer Amst Neth
2008
;
59
:
105
10
.
140.
Kim
D
,
Kim
H
,
Koo
JS
. 
Expression of caveolin-1, caveolin-2 and caveolin-3 in thyroid cancer and stroma
.
Pathobiology
2012
;
79
:
1
10
.
141.
Qian
X-L
,
Pan
Y-H
,
Huang
Q-Y
,
Shi
Y-B
,
Huang
Q-Y
,
Hu
Z-Z
, et al
Caveolin-1: a multifaceted driver of breast cancer progression and its application in clinical treatment
.
OncoTargets Ther
2019
;
12
:
1539
52
.
142.
Wang
S-W
,
Xu
K-L
,
Ruan
S-Q
,
Zhao
L-L
,
Chen
L-R
. 
Overexpression of caveolin-1 in cancer-associated fibroblasts predicts good outcome in breast cancer
.
Breast Care
2012
;
7
:
477
83
.
143.
Witkiewicz
AK
,
Dasgupta
A
,
Nguyen
KH
,
Liu
C
,
Kovatich
AJ
,
Schwartz
GF
, et al
Stromal caveolin-1 levels predict early DCIS progression to invasive breast cancer
.
Cancer Biol Ther
2009
;
8
:
1071
9
.
144.
Eliyatkin
N
,
Aktas
S
,
Diniz
G
,
Ozgur
HH
,
Ekin
ZY
,
Kupelioglu
A
. 
Expression of stromal caveolin- 1 may be a predictor for aggressive behaviour of breast cancer
.
Pathol Oncol Res
2018
;
24
:
59
65
.
145.
Qian
N
,
Ueno
T
,
Kawaguchi-Sakita
N
,
Kawashima
M
,
Yoshida
N
,
Mikami
Y
, et al
Prognostic significance of tumor/stromal caveolin-1 expression in breast cancer patients
.
Cancer Sci
2011
;
102
:
1590
6
.
146.
Chiu
W-T
,
Lee
H-T
,
Huang
F-J
,
Aldape
KD
,
Yao
J
,
Steeg
PS
, et al
Caveolin-1 upregulation mediates suppression of primary breast tumor growth and brain metastases by stat3 inhibition
.
Cancer Res
2011
;
71
:
4932
43
.
147.
Shi
Y
,
Tan
S-H
,
Ng
S
,
Zhou
J
,
Yang
N-D
,
Koo
G-B
, et al
Critical role of CAV1/caveolin-1 in cell stress responses in human breast cancer cells via modulation of lysosomal function and autophagy
.
Autophagy
2015
;
11
:
769
84
.
148.
Plaks
V
,
Koopman
CD
,
Werb
Z
. 
Circulating tumor cells
.
Science
2013
;
341
:
1186
8
.
149.
Beloribi-Djefaflia
S
,
Vasseur
S
,
Guillaumond
F
. 
Lipid metabolic reprogramming in cancer cells
.
Oncogenesis
2016
;
5
:
e189
.
150.
Gupta
VK
,
Banerjee
S
. 
Isolation of lipid raft proteins from CD133+ cancer stem cells
.
Methods Mol Biol
2017
;
1609
:
25
31
.
151.
Schworer
SA
,
Smirnova
II
,
Kurbatova
I
,
Bagina
U
,
Churova
M
,
Fowler
T
, et al
Toll-like receptor-mediated down-regulation of the deubiquitinase cylindromatosis (CYLD) protects macrophages from necroptosis in wild-derived mice
.
J Biol Chem
2014
;
289
:
14422
33
.
152.
He
S
,
Liang
Y
,
Shao
F
,
Wang
X
. 
Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3–mediated pathway
.
Proc Natl Acad Sci
2011
;
108
:
20054
9
.
153.
Yang
D
,
Liang
Y
,
Zhao
S
,
Ding
Y
,
Zhuang
Q
,
Shi
Q
, et al
ZBP1 mediates interferon-induced necroptosis
.
Cell Mol Immunol
2020
;
17
:
356
68
.
154.
Gu
L
,
Saha
ST
,
Thomas
J
,
Kaur
M
. 
Targeting cellular cholesterol for anticancer therapy
.
FEBS J
2019
;
286
:
4192
208
.
155.
Lee
SY
,
Ko
S-H
,
Shim
J-S
,
Kim
D-D
,
Cho
H-J
. 
Tumor targeting and lipid rafts disrupting hyaluronic acid-cyclodextrin-based nanoassembled structure for cancer therapy
.
ACS Appl Mater Interfaces
2018
;
10
:
36628
40
.