Despite recent advances in the treatment of hematologic malignancies, relapse still remains a consistent issue. One of the primary contributors to relapse is the bone marrow microenvironment providing a sanctuary to malignant cells. These cells interact with bone marrow components such as osteoblasts and stromal cells, extracellular matrix proteins, and soluble factors. These interactions, mediated by the cell surface proteins like cellular adhesion molecules (CAMs), induce intracellular signaling that leads to the development of bone marrow microenvironment–induced chemoprotection (BMC). Although extensive study has gone into these CAMs, including the development of targeted therapies, very little focus in hematologic malignancies has been put on a family of cell surface proteins that are just as important for mediating bone marrow interactions: the transmembrane 4 superfamily (tetraspanins; TSPANs). TSPANs are known to be important mediators of microenvironmental interactions and metastasis based on numerous studies in solid tumors. Recently, evidence of their possible role in hematologic malignancies, specifically in the regulation of cellular adhesion, bone marrow homing, intracellular signaling, and stem cell dynamics in malignant hematologic cells has come to light. Many of these effects are facilitated by associations with CAMs and other receptors on the cell surface in TSPAN-enriched microdomains. This could suggest that TSPANs play an important role in mediating BMC in hematologic malignancies and could be used as therapeutic targets. In this review, we discuss TSPAN structure and function in hematologic cells, their interactions with different cell surface and signaling proteins, and possible ways to target/inhibit their effects.

The bone marrow microenvironment (BMM) is the site of normal differentiation of hematopoietic stem cells (HSCs) and the development of healthy blood cells. Located within this microenvironment are also other cells such as osteoblasts, endothelial cells, stromal cells, and mesenchymal stem cells.1 During normal hematopoiesis, HSCs can interact with these cells or their secreted extracellular matrix proteins and soluble factors to foster proper development and differentiation.2 

Interaction of malignant cells with these cells and proteins can activate intracellular signaling pathways that drive cells into a chemoresistant state.3 Thus, association of cancer cells with BMM elements can be problematic in hematologic malignancies. This phenomenon is known as BMM–induced chemoprotection (BMC). Often, these effects rely on the expression of cellular adhesion molecules (CAMs) and their binding to cognate ligands.4 Therefore, the study of interactions between malignant cells and the BMM has been crucial to the better understanding of the development of hematologic malignancies and their resistance to chemotherapy.

Tetraspanins (TSPANs) are characterized by 4 transmembrane domains with intracellular N- and C-termini5 (Figure 1) and 2 extracellular loops, a short 13 to 30 amino acid loop (EC1) and a large 200 to 300 amino acid loop (EC2). TSPANs have not been shown to act like typical adhesion or ligand receptors; however, they can interact with other cell surface receptors, signaling molecules, and each other.6-8 There are 4 main domains, which have functional consequences within TSPANs9: (1) the EC2 loop contains a disulfide-stabilized hypervariable region and a highly conserved region,10 the variable region mediates the specific interactions unique to each TSPAN, whereas the conserved region is involved in homodimerization.11 (2) The transmembrane regions are important for the formation of the “TSPAN web” or “TSPAN-enriched microdomains” (TEMs),9 which will be discussed in more depth in further sections. These regions show a plurality of hydrophobic interactions, as well as strong hydrogen bonding between polarized amino acids of adjacent TSPAN neighbors, all of which allow for the tight packing and formation of TEMs.9 (3) Intracellularly, the C-terminus plays a role in intracellular signaling by interacting with different signaling molecules and cytoskeletal proteins.6,12-14 (4) TSPANs contain palmitoylation sites (typically cysteine residues modified by attachment of fatty acids), which are important for both signaling and the formation of new TEMs.15-18 

Figure 1.

Structure and domain functions of TSPANs. TSPANs are characterized by the presence of 4 transmembrane domains (1-4). There are multiple regions within the protein that mediate key effects. (A) Disulphide-stabilized (green spheres) variable region of the large extracellular loop (EC2) responsible for the unique interactions specific to each TSPAN. (B) The conserved region of the EC2 loop mediates homodimerization. (C) Transmembrane regions are important for the formation of TEMs. (D) Palmitoylation sites are crucial to the proper formation of new TEMs and play a role in signaling. (E) The C-terminus is responsible for interacting with signaling and cytoskeletal proteins.

Figure 1.

Structure and domain functions of TSPANs. TSPANs are characterized by the presence of 4 transmembrane domains (1-4). There are multiple regions within the protein that mediate key effects. (A) Disulphide-stabilized (green spheres) variable region of the large extracellular loop (EC2) responsible for the unique interactions specific to each TSPAN. (B) The conserved region of the EC2 loop mediates homodimerization. (C) Transmembrane regions are important for the formation of TEMs. (D) Palmitoylation sites are crucial to the proper formation of new TEMs and play a role in signaling. (E) The C-terminus is responsible for interacting with signaling and cytoskeletal proteins.

Close modal

TEMs, also called the TSPAN web, are dynamic membrane entities that act as signaling platforms.19 They are heterogeneous compositions of interconnected proteins that are involved in functionally related processes (Figure 2). TEMs consist of different TSPANs, adhesion molecules like integrins and immunoglobulin superfamily members, and other signaling receptors such as G-proteins and tyrosine kinases.20 TSPANs act like scaffolding proteins within these microdomains, by facilitating the assembly of these different proteins to potentiate the activation of associated intracellular signaling pathways.21-23 

Figure 2.

TEMs play a key role in TSPAN function. TEMs are dynamic membrane entities that play a key role in mediating interactions with the BM. TSPANs act as scaffolding proteins to bring together many proteins with similar functions such as CAMs (like integrins and immunoglobulin superfamily [IgSF] members) and signaling receptors (G protein–coupled receptors [GPCRs] and receptor tyrosine kinases [RTKs]). The crosslinking of TSPANs creates a large secondary signaling network, which can effectively transduce extracellular stimuli inside to intracellular signaling pathways.

Figure 2.

TEMs play a key role in TSPAN function. TEMs are dynamic membrane entities that play a key role in mediating interactions with the BM. TSPANs act as scaffolding proteins to bring together many proteins with similar functions such as CAMs (like integrins and immunoglobulin superfamily [IgSF] members) and signaling receptors (G protein–coupled receptors [GPCRs] and receptor tyrosine kinases [RTKs]). The crosslinking of TSPANs creates a large secondary signaling network, which can effectively transduce extracellular stimuli inside to intracellular signaling pathways.

Close modal

Within TEMs, TSPANs can have strong interactions with one another. Because of this, the specific interactions of each individual TSPAN can crosslink into large secondary signaling networks.19 This crosslinkage is stabilized by multiple regions within the TSPAN protein, such as the EC2 loop and the transmembrane domain but relies most heavily on the palmitoylation of intracellular cysteines.9 Despite their reliance on palmitoylation, TEMs reportedly stay intact after disruption of these palmitoylation sites; however, new TEMs were unable to form, suggesting that the palmitoylation is required for the initial formation.19 In addition to palmitoylation, TEMs also rely on the functional redundancy of other TSPAN domains such as the transmembrane regions and EC2 loops. These domains can interact with those on adjacent TSPAN proteins to allow for efficient formation and functional activity of TEMs.9 

The structure of TEMs is maintained by binding to cholesterol and sphingolipids, similar to that in lipid rafts, which are specialized microdomains that facilitate signal transduction.24 Although TEMs appear to be similar to lipid rafts, they are distinguished by extractability in strong detergents such as Triton X-100, and lack of glycosylphosphatidylinositol-anchored proteins.25 Recent studies have identified functional interactions between TEMs and lipid rafts mediated by TSPANs such as CD82.26 

TSPAN interactions with other cell surface proteins and the formation of TEMs are important for the function of a variety of cellular processes such as cellular adhesion, BM homing, survival signaling, and stem cell dynamics. Many of these functions have been discovered in solid tumors. However, evidence of TSPAN effects in hematologic cells and malignancies is growing. The effects of TSPANs on HSCs have been described earlier.27 In further sections, we provide specific examples of the aforementioned functions of TSPANs in hematologic cells, with an in-depth summary provided in Table 1.

Table 1.

Interactions and effects of TSPANs on malignant hematologic cells

ProteinCancerFunction/interactionRef
CD9 B-ALL Modulates CXCR-4–mediated cell migration through RAC1 signaling 28  
  Regulates LSC activity 29  
  Poor prognostic marker and blockade with anti-CD9 antibodies suppressed disease progression and enhanced chemosensitivity in vivo 30  
  Activates PI3K/Akt signaling via direct interaction with PI3K/p85 31  
  Mediates cellular adhesion and inhibits cellular death via a p53-dependent mechanism 32  
 B-CLL Abnormal expression associated with disease infiltration patterns 33  
 AML Relevant marker for MRD and targeting of LSCs 34  
 AML, CML, lymphoma Mediates self-renewal of LSCs via STAT5B signaling 35  
 Multiple myeloma Enhances susceptibility to cell-mediated cytolysis 36  
CD37 CLL Mediates prosurvival signaling via PI3K through ITAM-like motif in C-terminus, induces SHP1-dependent apoptotic signaling through ITIM-like motif in N-terminus 37  
 AML Poor prognostic marker 38  
CD53 Lymphoma Increases Bcl-XL and decreases bax expression via activation of Akt 39  
CD63 AML Expression on exosomes induces interleukin-8 production in bone marrow stromal cells to induce chemoprotection 40  
CD81 B-ALL Loss of expression associated with mature B-cells being ready to migrate and leave the BM 41  
  Mediates BMC and in vivo BM homing and engraftment, controls BTK signaling through CD19 expression, thereby inhibiting p53-mediated cell death 42  
 AML Controls cellular adhesion, migration, BM homing, and drug resistance 43  
 B-CLL Abnormal expression associated with disease infiltration patterns 33  
CD82 AML BM homing via N-cadherin organization 44  
  LSC adhesion and survival through STAT5/interleukin-10 45  
  Akt regulation of BCL2L12 46  
  Increases expression of EZH2 by inhibiting p38 activity 47  
  Activates β1 integrin and PKCα signaling 48  
  Promotes survival via activation of Wnt/β-catenin signaling 49  
 B-ALL Higher expression in malignant cells compared with nonmalignant cells, prognosis marker 50  
ProteinCancerFunction/interactionRef
CD9 B-ALL Modulates CXCR-4–mediated cell migration through RAC1 signaling 28  
  Regulates LSC activity 29  
  Poor prognostic marker and blockade with anti-CD9 antibodies suppressed disease progression and enhanced chemosensitivity in vivo 30  
  Activates PI3K/Akt signaling via direct interaction with PI3K/p85 31  
  Mediates cellular adhesion and inhibits cellular death via a p53-dependent mechanism 32  
 B-CLL Abnormal expression associated with disease infiltration patterns 33  
 AML Relevant marker for MRD and targeting of LSCs 34  
 AML, CML, lymphoma Mediates self-renewal of LSCs via STAT5B signaling 35  
 Multiple myeloma Enhances susceptibility to cell-mediated cytolysis 36  
CD37 CLL Mediates prosurvival signaling via PI3K through ITAM-like motif in C-terminus, induces SHP1-dependent apoptotic signaling through ITIM-like motif in N-terminus 37  
 AML Poor prognostic marker 38  
CD53 Lymphoma Increases Bcl-XL and decreases bax expression via activation of Akt 39  
CD63 AML Expression on exosomes induces interleukin-8 production in bone marrow stromal cells to induce chemoprotection 40  
CD81 B-ALL Loss of expression associated with mature B-cells being ready to migrate and leave the BM 41  
  Mediates BMC and in vivo BM homing and engraftment, controls BTK signaling through CD19 expression, thereby inhibiting p53-mediated cell death 42  
 AML Controls cellular adhesion, migration, BM homing, and drug resistance 43  
 B-CLL Abnormal expression associated with disease infiltration patterns 33  
CD82 AML BM homing via N-cadherin organization 44  
  LSC adhesion and survival through STAT5/interleukin-10 45  
  Akt regulation of BCL2L12 46  
  Increases expression of EZH2 by inhibiting p38 activity 47  
  Activates β1 integrin and PKCα signaling 48  
  Promotes survival via activation of Wnt/β-catenin signaling 49  
 B-ALL Higher expression in malignant cells compared with nonmalignant cells, prognosis marker 50  

TSPANs have a large variety of interacting partners and can mediate many different cellular processes.

B-CLL, B-cell chronic lymphocytic leukemia; BTK, Bruton tyrosine kinase; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; ITAM, immunoreceptor tyrosine-based activation motif; ITIM, immunoreceptor tyrosine-based inhibitory motif; LSC, leukemic stem cell; MRD, measurable residual disease; PKC, protein kinase Cα.

TSPANs are known for their ability to mediate interactions with the local microenvironment through their lateral associations with CAMs on the cell surface. One of the most common CAMs to interact with TSPANs is integrin α4β1 (VLA-4). When interacting with the TSPAN proteins CD9, CD81, or CD82, adhesion of VLA-4 to its target (vascular cell adhesion molecule-1) is increased.51-53 Often, this augmentation will occur via clustering of VLA-4 proteins on the cell surface. TSPAN-mediated adhesion can also be induced via activation of downstream signaling pathways associated with specific TSPAN proteins. Upon CD53 activation via monoclonal antibodies (mAbs) in lymphoma, phosphoinositide 3-kinase (PI3K) and protein kinase C signaling are activated intracellularly and lead to homotypic cellular adhesion.54 

CD82 is also known to mediate BM homing in acute myeloid leukemia (AML) via cell surface clustering of the CAM N-cadherin.44 This clustering is reliant upon the proper palmitoylation and N-glycosylation of CD82. CD82 palmitoylation mutants lack N-cadherin assembly into clusters whereas N-glycosylation mutants lack adequate nanoscale packing. Increased cellular adhesion in the BM in hematologic malignancies can lead to the activation of chemoprotective signaling. Therefore, TSPAN functions can be indirectly responsible for mediating BMC. In AML, CD82-mediated adhesion enhances the activation of STAT5/Akt signaling, culminating in the increased expression of the antiapoptotic protein BCL2L12 (Figure 3).45,46 Considering the role of CD82 in mediating cellular adhesion to the BM via cell surface clustering of N-cadherin, it is possible that these signaling events are downstream of N-cadherin. However, this still requires further study.

Figure 3.

CD82 mediates leukemic stem cell survival and interactions with the microenvironment. CD82 mediates leukemic stem cell (LSC) interaction with the BM in multiple ways. It increases microenvironment interaction by organizing N-cadherin at the cell surface in high densities via CD82 palmitoylation (green triangles) and glycosylation (red spheres). However, the connection of N-cadherin to CD82-associated intracellular effects is still unknown. CD82-associated STAT5 signaling can combine with increased p-Akt (via inhibition of PTEN by DNA methylation from EZH2 due to downregulation of p38 caused by CD82-associated signaling) to increase the expression of antiapoptotic protein BCL2L12. The combination of these effects leads to increased LSC survival.

Figure 3.

CD82 mediates leukemic stem cell survival and interactions with the microenvironment. CD82 mediates leukemic stem cell (LSC) interaction with the BM in multiple ways. It increases microenvironment interaction by organizing N-cadherin at the cell surface in high densities via CD82 palmitoylation (green triangles) and glycosylation (red spheres). However, the connection of N-cadherin to CD82-associated intracellular effects is still unknown. CD82-associated STAT5 signaling can combine with increased p-Akt (via inhibition of PTEN by DNA methylation from EZH2 due to downregulation of p38 caused by CD82-associated signaling) to increase the expression of antiapoptotic protein BCL2L12. The combination of these effects leads to increased LSC survival.

Close modal

TSPANs also play a role in mediating the expression of key surface receptor components such as CD19, which is part of the B-cell coreceptor. CD19 relies on the expression of CD81 for proper trafficking to the cell membrane.41 CD81 downregulation was observed in B-cell acute lymphoblastic (B-ALL) cells resistant to blinatumomab, which targets CD19, indicating that cancer cells disrupt this association as a mechanism to overcome blinatumomab sensitivity.55 Likewise, in B-ALL CD81-knockout cells, CD19 expression was completely lost on the cell surface. This resulted in the depletion of Bruton tyrosine kinase signaling and a sensitization to chemotherapy.42 This highlights the secondary effect that TSPANs can have on intracellular signaling by controlling the expression of specific cell surface proteins.

TSPANs can also directly initiate signaling pathways via their intracellular N- and C-termini. Antibody ligation of CD37 using a mAb in chronic lymphocytic leukemia can activate 2 separate, contradictory signaling pathways (Figure 4). After ligation, an N-terminus immunoreceptor tyrosine-based inhibitory motif–like motif becomes phosphorylated and forms a SHP1 phosphatase–containing complex that inhibits Akt and induces cell death. However, ligation also phosphorylates a C-terminus immunoreceptor tyrosine-based activation motif–like motif, which mediates prosurvival signaling through PI3Kδ activity.37 Therefore, it is possible that activation of the immunoreceptor tyrosine-based activation motif–like motif via lateral surface associations and interactions within the BMM could play a critical role in inducing malignant cell survival. However, because of the cell death properties associated with the immunoreceptor tyrosine-based inhibitory motif–like motif, combination therapy treatment of anti-CD37 mAb with PI3Kδ inhibitors could provide a synergistic mechanism for inducing cancer cell death.

Figure 4.

CD37 mediates both prosurvival and prodeath effects from its N- and C-termini. CD37 initiates cell death via phosphorylation of an immune tyrosine-based inhibitory motif (ITIM)-like motif in its N-terminus tail. This leads to a SHP1-dependent cell death. IT can also initiate prosurvival signaling through phosphorylation of its immune tyrosine-based activation motif (ITAM) on the C-terminus tail. This occurs through PI3K/Akt signaling.

Figure 4.

CD37 mediates both prosurvival and prodeath effects from its N- and C-termini. CD37 initiates cell death via phosphorylation of an immune tyrosine-based inhibitory motif (ITIM)-like motif in its N-terminus tail. This leads to a SHP1-dependent cell death. IT can also initiate prosurvival signaling through phosphorylation of its immune tyrosine-based activation motif (ITAM) on the C-terminus tail. This occurs through PI3K/Akt signaling.

Close modal

The presence of stem cell properties is a major hallmark of malignant cells, and TSPANs play an important role in regulating these processes. CD9 is known to mediate the stem cell properties of B-ALL cells via modulation of the expression of Src family proteins and the upregulation of the histone deubiquitinase USP22.29 It is also a modulator of STAT5B signaling, which is responsible for HSC and leukemic stem cell self-renewal.35 Although the study of TSPANs and their regulation of stem cell dynamics in hematologic malignancies still requires further investigation, their role in HSCs has been better defined. Expression of CD81 is crucial for the maintenance of long-term HSCs via its deactivation of the Akt pathway after proliferative stress.56 This allows for a return to a quiescent state, a state that has therapeutic impacts in malignant cells. CD82 can also maintain this quiescent state in HSCs when it binds to its partner CD234 (DARC) on BM macrophages.57 This interaction stabilizes CD82 on the cell surface, thus activating protein kinase Cα–dependent TGF-β1/Smad3 signaling, resulting in cell cycle inhibition.

It is evident how TSPANs can control the cell surface dynamics and microenvironmental interactions of hematologic malignant cells. In addition to these phenomena, TSPANs also participate in intercellular communication as mediators of extracellular vesicle formation (exosomes) and are considered as markers for exosomes.58 TSPANs such as CD9, CD63, and CD81 play a critical role in cargo and target selection of exosomes, which can have significant effects on cancer progression.59 Cross talk between AML cells and BM mesenchymal stem cells mediated by AML-derived exosomes induce interleukin 8 secretion from mesenchymal stem cells. This results in a decreased sensitivity of AML cells to etoposide treatment.40 Although it was reported that the exosomes secreted by AML cells expressed TSPAN CD63, it remains to be determined whether CD63 expression promoted chemoresistance, as seen in the case of CD9-bearing exosomes that promote cancer cell migration.60 

Malignant cell–derived exosomes can also modulate the organizational makeup of the BM. After exosome release from chronic lymphocytic leukemia cells, stromal cells underwent a transition into cancer-associated fibroblasts, whose increased cytokine secretions favored malignant cell growth.61 Induction of angiogenesis can also result from exosome release and favor the growth and survival of hematologic malignancies. In multiple myeloma, exosomes containing miR-135b were identified to be the mediators of factor-inhibiting hypoxia-inducible factor 1 expression and subsequent angiogenic formations.62 

Immune evasion/suppression is another key aspect of malignant cell survival and progression. Exosomes derived from tumor cells are known to expand regulatory T-cell populations, thus preventing immune cell targeting of leukemic cells.63,64 It is intriguing to note that CD81 expression has been identified as a critical regulator of regulatory T-cell function.65 Because of this and the prominent expression of TSPANs on exosomes, it is possible that TSPANs could play a large role in cancer progression via immunosuppression. However, further investigation is still required.

TSPANs are potent mediators of BM interactions and cell signaling. Because of these effects, TSPANs make good therapeutic targets to improve treatment efficacy in hematologic malignancies (Figure 5).

Figure 5.

Possible methods to target TSPANs. There have been multiple proposed mechanisms on how to target TSPANs and inhibit their functions. (A) mAbs can be used to antagonize TSPANs either directly, or by inhibiting its lateral interactions. (B) Soluble EC2 loops can be used to disrupt the unique interactions of each TSPAN. (C) Analogues to transmembrane regions can prevent the formation of TEMs, thereby disrupting their effects. (D) Inhibiting palmitoylation can also be used as a mechanism to prevent TEM formation. (E) Many TSPANs have a motif in their C-terminus that can interact with PDZ domains on different signaling proteins. Small molecule inhibitors to these domains can prevent their activation, and therefore the cellular responses.

Figure 5.

Possible methods to target TSPANs. There have been multiple proposed mechanisms on how to target TSPANs and inhibit their functions. (A) mAbs can be used to antagonize TSPANs either directly, or by inhibiting its lateral interactions. (B) Soluble EC2 loops can be used to disrupt the unique interactions of each TSPAN. (C) Analogues to transmembrane regions can prevent the formation of TEMs, thereby disrupting their effects. (D) Inhibiting palmitoylation can also be used as a mechanism to prevent TEM formation. (E) Many TSPANs have a motif in their C-terminus that can interact with PDZ domains on different signaling proteins. Small molecule inhibitors to these domains can prevent their activation, and therefore the cellular responses.

Close modal

One way of targeting TSPANs is the use of mAbs, which have been widely used against CAMs in hematologic malignancies to improve outcomes.66 In most cases, these mAbs prevent a specific interaction between CAMs and their targets, either on other cells or in the ECM. mAbs against TSPANs can be antagonistic by diminishing associated cellular functions, either directly or by blocking lateral interactions (Table 2). For example, a unique CD81-targeting mAb that promotes CD81 clustering reportedly induces cytotoxicity in lymphoma cells via direct killing or via activation of the innate immune system.67,68 CD82-blocking mAbs potentiated the effect of cytarabine by disrupting AML cell adhesion to the BMM and improved survival in mice with AML cell engraftment.69 CD37 mAbs displayed proapoptotic effects in B-cell malignancies.70 A bifunctional mAb pair recognizing CD37 and CD20 mixed in a 1:1 ratio (PSB202)71 is currently being evaluated in the clinic. Moreover, CD37-targeting mAbs have been used in combination with PI3K inhibitors in preclinical studies to improve efficacy in chronic lymphocytic leukemia.72,73 

Table 2.

Current TSPAN-targeting therapies in hematologic malignancies

TherapeuticTarget and effectsModelReference
Anti-CD82 CD82: mobilizing agent, potentiates effects of anticancer therapy Preclinical 69  
5A6 CD81: mAb selectively kills B-cell lymphoma cells via direct cell death and innate immune system activation Preclinical 67  
PSB202 CD37-CD20: bifunctional mAb pair to enable direct killing of B cells in lymphomas Clinical NCT05003141 
CAR-37 CD37: CAR T cells for CD37+ hematologic malignancies Clinical NCT04136275 
TherapeuticTarget and effectsModelReference
Anti-CD82 CD82: mobilizing agent, potentiates effects of anticancer therapy Preclinical 69  
5A6 CD81: mAb selectively kills B-cell lymphoma cells via direct cell death and innate immune system activation Preclinical 67  
PSB202 CD37-CD20: bifunctional mAb pair to enable direct killing of B cells in lymphomas Clinical NCT05003141 
CAR-37 CD37: CAR T cells for CD37+ hematologic malignancies Clinical NCT04136275 

CAR, chimeric antigen receptor.

Although mAbs offer a simple way to target TSPANs, it is not as simplistic as it may sound. Although many TSPANs are expressed at high levels in hematologic malignancies, they are also expressed on a variety of cell types throughout the body, both hematologic and nonhematologic, leading to off-target effects. However, studies using transgenic mice have identified that, despite knock out of these proteins, mice generally avoid overly serious side effects (Table 3). Encouragingly, targeting CD81 using 5A6 mAb selectively killed lymphoma cells while sparing normal hematopoietic cells, likely because of the higher CD81 expression in lymphoma.67 Another issue is that it can be difficult to know which cellular functions may be affected by using a mAb targeting a specific epitope. TSPANs can mediate a multitude of different processes, therefore, it can be hard to discern whether an antagonistic or agonistic response against a specific biologic process will be achieved.74,75 

Table 3.

Physiologic effects of different TSPAN knockouts in mouse models

ProteinKO phenotypePhysiologic consequenceReference
CD9 Enhanced lung inflammation Reduced response to statins 76  
 Decreased lymphangiogenesis Abnormal development of lymphatic vasculature 77  
CD37 Deficiencies in T-cell–dependent B-cell responses Reduced antibody responses to antigens 78  
 Dysregulation of TCR signaling Hyperproliferative T cells 79  
 Increased antigen presentation by dendritic cells Hyperstimulation of T cells 80  
 Impaired neutrophil recruitment Decreased inflammatory response 81  
CD63 Significant reduction in leukocyte rolling, recruitment, and extravasation Decreased inflammatory response 82  
CD81 Lower expression of CD19 on B cells, leading to lower Ca2+ mobilization after CD19 stimulation Abnormal B-cell antibody responses 83  
 Reduced production of IgG1 in Th2 immune responses Reduced B1 lymphocyte response 84  
 Decreased lymphangiogenesis Abnormal development of lymphatic vasculature 77  
CD151 Defects in hemostasis and in αIIbβ3 integrin signaling Disrupted platelet function 85  
 Hyperproliferation of T cells in response to stimulation with mitogenic agents Cellular hypersensitivity to activation 86  
ProteinKO phenotypePhysiologic consequenceReference
CD9 Enhanced lung inflammation Reduced response to statins 76  
 Decreased lymphangiogenesis Abnormal development of lymphatic vasculature 77  
CD37 Deficiencies in T-cell–dependent B-cell responses Reduced antibody responses to antigens 78  
 Dysregulation of TCR signaling Hyperproliferative T cells 79  
 Increased antigen presentation by dendritic cells Hyperstimulation of T cells 80  
 Impaired neutrophil recruitment Decreased inflammatory response 81  
CD63 Significant reduction in leukocyte rolling, recruitment, and extravasation Decreased inflammatory response 82  
CD81 Lower expression of CD19 on B cells, leading to lower Ca2+ mobilization after CD19 stimulation Abnormal B-cell antibody responses 83  
 Reduced production of IgG1 in Th2 immune responses Reduced B1 lymphocyte response 84  
 Decreased lymphangiogenesis Abnormal development of lymphatic vasculature 77  
CD151 Defects in hemostasis and in αIIbβ3 integrin signaling Disrupted platelet function 85  
 Hyperproliferation of T cells in response to stimulation with mitogenic agents Cellular hypersensitivity to activation 86  

Rargeting TSPANs can be difficult because of their widespread expression on many cell types; despite these complications, many of the effects of TSPAN-knockout models can be mild, lending support to the development of anti-TSPAN therapies.

IgG, immunoglobulin G; TCR, T-cell receptor; Th2, T helper 2 cell.

Soluble EC2 loops are another way to target TSPANs by disrupting the specific interactions of each TSPAN protein.87 The lateral effects of TSPANs, like inducing VLA-4 binding to vascular cell adhesion molecule-1, can be diminished by this approach, thereby inhibiting responses to microenvironmental support. EC2 loops for CD9 and CD151 have previously been used to prevent extravasation by blocking endothelial cell interactions with leukocytes.88 

The transmembrane regions of TSPANs are important for the development and stability of TEMs, which are a key part of the effects of TSPANs. Although no current studies have identified their effectiveness, structural analogues to these transmembrane regions have been proposed to be a possible way to disrupt TEM formation.89 

Palmitoylation is critical for the formation of new TEMs; therefore, targeting TSPAN palmitoylation can also be used as a mechanism to prevent TEM formation. One way to achieve this is by inhibiting the activity of protein acyl transferases such as DHHC2, which reportedly affect the palmitoylation and stability of CD9 and CD151.90 

Targeting the C-terminus can prevent the downstream effects of TSPANs. Specifically, many TSPANs have a motif in their C-terminus that can bind to PDZ domains present on cytoskeletal signaling proteins such as syntenin-1.91 Small-molecule inhibitors to these PDZ domains can prevent the transduction of extracellular stimuli into the cell, and prevent the corresponding cellular responses.92 

Although most of the aforementioned targeting mechanisms focus on inhibiting TSPAN functions, some effects of TSPANs can be beneficial, and therefore, should be encouraged. This can be achieved by either adenoviral transductions or via targeted knock down of proteins known to mediate TSPAN degradation, such as gp78 ubiquitin ligase, which degrades CD82.93,94 Furthermore, TSPANs like CD81 and CD82 can have negative effects on cell survival and invasiveness in hematologic malignancies like multiple myeloma.95 Treatment with hypomethylating agents such 5-aza-2′-deoxycytidine can potentially elevate their expression by desilencing these genes and show a positive outcome.

TSPANs can be targeted in a multitude of ways, although many of the therapies are still in their infancy, especially in hematologic malignancies. If more targeted therapies can be successfully developed, they could go a long way in improving the overall efficacy of treatment.

It is worth noting that, although TSPANs have not been identified to act as typical receptors and have no known cell-to-cell binding partners, it is possible that TSPANs could participate in homophilic or heterophilic binding via their EC2 loops. As mentioned earlier, the conserved region of the EC2 loop is responsible for mediating lateral homodimerization within the cell membrane9; however, it may be possible that these effects are seen in cell-to-cell contact as well. Moreover, the variable region of the EC2 loop dictates the specific interactome of each TSPAN. This region could possibly also mediate cell-to-cell contact by binding with TSPANs on other cells, or with other CAMs such as integrins through arginine-glycine-aspartic acid domains, which are known to interact with TSPANs.96 The BM plays a critical role in the progression and persistence of hematologic malignancies. By complexing with many different CAMs such as integrins and immunoglobulin superfamily members, and by using their ability to form dynamic membrane signaling entities (TEMs), TSPANs are potentially key mediators of interactions within the BM. Although different targeted therapies developed against individual CAMs have achieved moderate success, targeting TSPANs instead or in addition to individual CAMs could provide more comprehensive success. Further studies are needed not only to understand the role of TSPANs in hematologic malignancies but also in developing robust therapies that can target them and mitigate their effects.

The authors gratefully acknowledge the funding support from the Andrew McDonough B+ Foundation, the Lisa Dean Moseley Foundation, the Leukemia Research Foundation of Delaware, and the Nemours Foundation.

Contribution: A.Q. researched and developed first draft and figures; A.G. conceptualized and edited draft manuscript; and S.P.B. conceptualized, researched, and edited the draft of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

The current affiliation for A.Q. is Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL.

Correspondence: Sonali P. Barwe, Nemours Children’s Hospital, Rm 5218, E400 Dupont Experimental Station, 200 Powder Mill Rd, Wilmington, DE 19803; e-mail: sbarwe@nemours.org.

1.
Sidhu
I
,
Barwe
SP
,
Gopalakrishnapillai
A
.
The extracellular matrix: a key player in the pathogenesis of hematologic malignancies
.
Blood Rev
.
2021
. ;
48
:
100787
.
2.
Birbrair
A
,
Frenette
PS
.
Niche heterogeneity in the bone marrow
.
Ann N Y Acad Sci
.
2016
. ;
1370
(
1
):
82
-
96
.
3.
Meads
MB
,
Hazlehurst
LA
,
Dalton
WS
.
The bone marrow microenvironment as a tumor sanctuary and contributor to drug resistance
.
Clin Cancer Res
.
2008
. ;
14
(
9
):
2519
-
2526
.
4.
Barwe
SP
,
Quagliano
A
,
Gopalakrishnapillai
A
.
Eviction from the sanctuary: development of targeted therapy against cell adhesion molecules in acute lymphoblastic leukemia
.
Semin Oncol
.
2017
. ;
44
(
2
):
101
-
112
.
5.
Horejsi
V
,
Vlcek
C
.
Novel structurally distinct family of leucocyte surface glycoproteins including CD9, CD37, CD53 and CD63
.
FEBS Lett
.
1991
. ;
288
(
1-2
):
1
-
4
.
6.
Termini
CM
,
Gillette
JM
.
Tetraspanins function as regulators of cellular signaling
.
Front Cell Dev Biol
.
2017
. ;
5
:
34
.
7.
Berditchevski
F
,
Odintsova
E
.
Characterization of integrin-tetraspanin adhesion complexes: role of tetraspanins in integrin signaling
.
J Cell Biol
.
1999
. ;
146
(
2
):
477
-
492
.
8.
Charrin
S
,
Jouannet
S
,
Boucheix
C
,
Rubinstein
E
.
Tetraspanins at a glance
.
J Cell Sci
.
2014
. ;
127
(
pt 17
):
3641
-
3648
.
9.
Stipp
CS
,
Kolesnikova
TV
,
Hemler
ME
.
Functional domains in tetraspanin proteins
.
Trends Biochem Sci
.
2003
. ;
28
(
2
):
106
-
112
.
10.
Seigneuret
M
,
Delaguillaumie
A
,
Lagaudriere-Gesbert
C
,
Conjeaud
H
.
Structure of the tetraspanin main extracellular domain. A partially conserved fold with a structurally variable domain insertion
.
J Biol Chem
.
2001
. ;
276
(
43
):
40055
-
40064
.
11.
Kitadokoro
K
,
Bordo
D
,
Galli
G
, et al
.
CD81 extracellular domain 3D structure: insight into the tetraspanin superfamily structural motifs
.
EMBO J
.
2001
. ;
20
(
1-2
):
12
-
18
.
12.
Berditchevski
F
,
Tolias
KF
,
Wong
K
,
Carpenter
CL
,
Hemler
ME
.
A novel link between integrins, transmembrane-4 superfamily proteins (CD63 and CD81), and phosphatidylinositol 4-kinase
.
J Biol Chem
.
1997
. ;
272
(
5
):
2595
-
2598
.
13.
Carloni
V
,
Mazzocca
A
,
Ravichandran
KS
.
Tetraspanin CD81 is linked to ERK/MAPKinase signaling by Shc in liver tumor cells
.
Oncogene
.
2004
. ;
23
(
8
):
1566
-
1574
.
14.
Delaguillaumie
A
,
Lagaudriere-Gesbert
C
,
Popoff
MR
,
Conjeaud
H
.
Rho GTPases link cytoskeletal rearrangements and activation processes induced via the tetraspanin CD82 in T lymphocytes
.
J Cell Sci
.
2002
. ;
115
(
pt 2
):
433
-
443
.
15.
Berditchevski
F
,
Odintsova
E
,
Sawada
S
,
Gilbert
E
.
Expression of the palmitoylation-deficient CD151 weakens the association of alpha 3 beta 1 integrin with the tetraspanin-enriched microdomains and affects integrin-dependent signaling
.
J Biol Chem
.
2002
. ;
277
(
40
):
36991
-
37000
.
16.
Charrin
S
,
Manie
S
,
Oualid
M
,
Billard
M
,
Boucheix
C
,
Rubinstein
E
.
Differential stability of tetraspanin/tetraspanin interactions: role of palmitoylation
.
FEBS Lett
.
2002
. ;
516
(
1-3
):
139
-
144
.
17.
Yang
XH
,
Kovalenko
OV
,
Kolesnikova
TV
, et al
.
Contrasting effects of EWI proteins, integrins, and protein palmitoylation on cell surface CD9 organization
.
J Biol Chem
.
2006
. ;
281
(
18
):
12976
-
12985
.
18.
Delandre
C
,
Penabaz
TR
,
Passarelli
AL
,
Chapes
SK
,
Clem
RJ
.
Mutation of juxtamembrane cysteines in the tetraspanin CD81 affects palmitoylation and alters interaction with other proteins at the cell surface
.
Exp Cell Res
.
2009
. ;
315
(
11
):
1953
-
1963
.
19.
Hemler
ME
.
Tetraspanin functions and associated microdomains
.
Nat Rev Mol Cell Biol
.
2005
. ;
6
(
10
):
801
-
811
.
20.
Yanez-Mo
M
,
Barreiro
O
,
Gordon-Alonso
M
,
Sala-Valdes
M
,
Sanchez-Madrid
F
.
Tetraspanin-enriched microdomains: a functional unit in cell plasma membranes
.
Trends Cell Biol
.
2009
. ;
19
(
9
):
434
-
446
.
21.
Barreiro
O
,
Zamai
M
,
Yanez-Mo
M
, et al
.
Endothelial adhesion receptors are recruited to adherent leukocytes by inclusion in preformed tetraspanin nanoplatforms
.
J Cell Biol
.
2008
. ;
183
(
3
):
527
-
542
.
22.
Tarrant
JM
,
Robb
L
,
van Spriel
AB
,
Wright
MD
.
Tetraspanins: molecular organisers of the leukocyte surface
.
Trends Immunol
.
2003
. ;
24
(
11
):
610
-
617
.
23.
van Deventer
SJ
,
Dunlock
VME
,
van Spriel
AB
.
Molecular interactions shaping the tetraspanin web
.
Biochem Soc Trans
.
2017
. ;
45
(
3
):
741
-
750
.
24.
Odintsova
E
,
Butters
TD
,
Monti
E
,
Sprong
H
,
van Meer
G
,
Berditchevski
F
.
Gangliosides play an important role in the organization of CD82-enriched microdomains
.
Biochem J
.
2006
. ;
400
(
2
):
315
-
325
.
25.
Hemler
ME
.
Tetraspanin proteins mediate cellular penetration, invasion, and fusion events and define a novel type of membrane microdomain
.
Annu Rev Cell Dev Biol
.
2003
. ;
19
:
397
-
422
.
26.
Huang
C
,
Hays
FA
,
Tomasek
JJ
,
Benyajati
S
,
Zhang
XA
.
Tetraspanin CD82 interaction with cholesterol promotes extracellular vesicle-mediated release of ezrin to inhibit tumour cell movement
.
J Extracell Vesicles
.
2020
. ;
9
(
1
):
1692417
.
27.
Balise
VD
,
Saito-Reis
CA
,
Gillette
JM
.
Tetraspanin scaffold proteins function as key regulators of hematopoietic stem cells
.
Front Cell Dev Biol
.
2020
. ;
8
:
598
.
28.
Arnaud
MP
,
Vallee
A
,
Robert
G
, et al
.
CD9, a key actor in the dissemination of lymphoblastic leukemia, modulating CXCR4-mediated migration via RAC1 signaling
.
Blood
.
2015
. ;
126
(
15
):
1802
-
1812
.
29.
Yamazaki
H
,
Xu
CW
,
Naito
M
, et al
.
Regulation of cancer stem cell properties by CD9 in human B-acute lymphoblastic leukemia
.
Biochem Biophys Res Commun
.
2011
. ;
409
(
1
):
14
-
21
.
30.
Leung
KT
,
Zhang
C
,
Chan
KYY
, et al
.
CD9 blockade suppresses disease progression of high-risk pediatric B-cell precursor acute lymphoblastic leukemia and enhances chemosensitivity
.
Leukemia
.
2020
. ;
34
(
3
):
709
-
720
.
31.
Shi
YF
,
Huang
ZY
,
Huang
YS
, et al
.
Interaction between CD9 and PI3K-p85 activates the PI3K/AKT signaling pathway in B-lineage acute lymphoblastic leukemia
.
Oncol Rep
.
2021
. ;
46
(
1
):
140
.
32.
Xing
C
,
Xu
W
,
Shi
Y
, et al
.
CD9 knockdown suppresses cell proliferation, adhesion, migration and invasion, while promoting apoptosis and the efficacy of chemotherapeutic drugs and imatinib in Ph+ ALL SUP-B15 cells
.
Mol Med Rep
.
2020
. ;
22
(
4
):
2791
-
2800
.
33.
Barrena
S
,
Almeida
J
,
Yunta
M
, et al
.
Aberrant expression of tetraspanin molecules in B-cell chronic lymphoproliferative disorders and its correlation with normal B-cell maturation
.
Leukemia
.
2005
. ;
19
(
8
):
1376
-
1383
.
34.
Touzet
L
,
Dumezy
F
,
Roumier
C
, et al
.
CD9 in acute myeloid leukemia: prognostic role and usefulness to target leukemic stem cells
.
Cancer Med
.
2019
. ;
8
(
3
):
1279
-
1288
.
35.
Kollmann
S
,
Grausenburger
R
,
Klampfl
T
, et al
.
A STAT5B-CD9 axis determines self-renewal in hematopoietic and leukemic stem cells
.
Blood
.
2021
. ;
138
(
23
):
2347
-
2359
.
36.
Shallal
S
,
Kornbluth
J
.
CD9 expression enhances the susceptibility of myeloma cell lines to cell-mediated cytolysis
.
Blood
.
2000
. ;
96
(
1
):
224
-
233
.
37.
Lapalombella
R
,
Yeh
YY
,
Wang
L
, et al
.
Tetraspanin CD37 directly mediates transduction of survival and apoptotic signals
.
Cancer Cell
.
2012
. ;
21
(
5
):
694
-
708
.
38.
Zhang
Q
,
Han
Q
,
Zi
J
,
Song
C
,
Ge
Z
.
CD37 high expression as a potential biomarker and association with poor outcome in acute myeloid leukemia
.
Biosci Rep
.
2020
. ;
40
(
5
):
BSR20200008
.
39.
Yunta
M
,
Lazo
PA
.
Apoptosis protection and survival signal by the CD53 tetraspanin antigen
.
Oncogene
.
2003
. ;
22
(
8
):
1219
-
1224
.
40.
Chen
T
,
Zhang
G
,
Kong
L
,
Xu
S
,
Wang
Y
,
Dong
M
.
Leukemia-derived exosomes induced IL-8 production in bone marrow stromal cells to protect the leukemia cells against chemotherapy
.
Life Sci
.
2019
. ;
221
:
187
-
195
.
41.
Shoham
T
,
Rajapaksa
R
,
Boucheix
C
, et al
.
The tetraspanin CD81 regulates the expression of CD19 during B cell development in a postendoplasmic reticulum compartment
.
J Immunol
.
2003
. ;
171
(
8
):
4062
-
4072
.
42.
Quagliano
A
,
Gopalakrishnapillai
A
,
Kolb
EA
,
Barwe
S
.
Modulation of CD81 by epigenetic drug combination sensitizes acute lymphoblastic leukemia via decreased BTK signaling
.
Blood
.
2019
. ;
134
(
suppl 1
). 2628-2628.
43.
Gonzales
F
,
Boyer
T
,
Peyrouze
P
, et al
.
Targeting aberrant expression of CD81 impacts cell adhesion and migration, drug resistance and prognosis of acute myeloid leukemia
.
Blood
.
2017
. ;
130
(
suppl 1
). 2675-2675.
44.
Marjon
KD
,
Termini
CM
,
Karlen
KL
, et al
.
Tetraspanin CD82 regulates bone marrow homing of acute myeloid leukemia by modulating the molecular organization of N-cadherin
.
Oncogene
.
2016
. ;
35
(
31
):
4132
-
4140
.
45.
Nishioka
C
,
Ikezoe
T
,
Yang
J
, et al
.
CD82 regulates STAT5/IL-10 and supports survival of acute myelogenous leukemia cells
.
Int J Cancer
.
2014
. ;
134
(
1
):
55
-
64
.
46.
Nishioka
C
,
Ikezoe
T
,
Takeuchi
A
,
Nobumoto
A
,
Tsuda
M
,
Yokoyama
A
.
The novel function of CD82 and its impact on BCL2L12 via AKT/STAT5 signal pathway in acute myelogenous leukemia cells
.
Leukemia
.
2015
. ;
29
(
12
):
2296
-
2306
.
47.
Nishioka
C
,
Ikezoe
T
,
Yang
J
,
Yokoyama
A
.
Tetraspanin family member, CD82, regulates expression of EZH2 via inactivation of p38 MAPK signaling in leukemia cells
.
PLoS One
.
2015
. ;
10
(
5
):
e0125017
.
48.
Floren
M
,
Restrepo Cruz
S
,
Termini
CM
,
Marjon
KD
,
Lidke
KA
,
Gillette
JM
.
Tetraspanin CD82 drives acute myeloid leukemia chemoresistance by modulating protein kinase C alpha and beta1 integrin activation
.
Oncogene
.
2020
. ;
39
(
19
):
3910
-
3925
.
49.
Ji
H
,
Chen
L
,
Xing
Y
, et al
.
CD82 supports survival of childhood acute myeloid leukemia cells via activation of Wnt/beta-catenin signaling pathway
.
Pediatr Res
.
2019
. ;
85
(
7
):
1024
-
1031
.
50.
Ji
H
,
Chen
L
,
Dai
Y
, et al
.
Aberrant expression of CD133 and CD82 in patients with pediatric acute lymphoblastic leukemia and the clinical significance
.
Oncol Lett
.
2017
. ;
14
(
5
):
5811
-
5818
.
51.
Yoon
SO
,
Lee
IY
,
Zhang
X
,
Zapata
MC
,
Choi
YS
.
CD9 may contribute to the survival of human germinal center B cells by facilitating the interaction with follicular dendritic cells
.
FEBS Open Bio
.
2014
. ;
4
:
370
-
376
.
52.
Spring
FA
,
Griffiths
RE
,
Mankelow
TJ
, et al
.
Tetraspanins CD81 and CD82 facilitate alpha4beta1-mediated adhesion of human erythroblasts to vascular cell adhesion molecule-1
.
PLoS One
.
2013
. ;
8
(
5
):
e62654
.
53.
Saito-Reis
CA
,
Marjon
KD
,
Pascetti
EM
,
Floren
M
,
Gillette
JM
.
The tetraspanin CD82 regulates bone marrow homing and engraftment of hematopoietic stem and progenitor cells
.
Mol Biol Cell
.
2018
. ;
29
(
24
):
2946
-
2958
.
54.
Lazo
PA
,
Cuevas
L
,
Gutierrez del Arroyo
A
,
Orue
E
.
Ligation of CD53/OX44, a tetraspan antigen, induces homotypic adhesion mediated by specific cell-cell interactions
.
Cell Immunol
.
1997
. ;
178
(
2
):
132
-
140
.
55.
Braig
F
,
Brandt
A
,
Goebeler
M
, et al
.
Resistance to anti-CD19/CD3 BiTE in acute lymphoblastic leukemia may be mediated by disrupted CD19 membrane trafficking
.
Blood
.
2017
. ;
129
(
1
):
100
-
104
.
56.
Lin
KK
,
Rossi
L
,
Boles
NC
,
Hall
BE
,
George
TC
,
Goodell
MA
.
CD81 is essential for the re-entry of hematopoietic stem cells to quiescence following stress-induced proliferation via deactivation of the Akt pathway
.
PLoS Biol
.
2011
. ;
9
(
9
):
e1001148
.
57.
Hur
J
,
Choi
JI
,
Lee
H
, et al
.
CD82/KAI1 maintains the dormancy of long-term hematopoietic stem cells through interaction with DARC-expressing macrophages
.
Cell Stem Cell
.
2016
. ;
18
(
4
):
508
-
521
.
58.
Andreu
Z
,
Yanez-Mo
M
.
Tetraspanins in extracellular vesicle formation and function
.
Front Immunol
.
2014
. ;
5
:
442
.
59.
Malla
RR
,
Pandrangi
S
,
Kumari
S
,
Gavara
MM
,
Badana
AK
.
Exosomal tetraspanins as regulators of cancer progression and metastasis and novel diagnostic markers
.
Asia Pac J Clin Oncol
.
2018
. ;
14
(
6
):
383
-
391
.
60.
Miki
Y
,
Yashiro
M
,
Okuno
T
, et al
.
CD9-positive exosomes from cancer-associated fibroblasts stimulate the migration ability of scirrhous-type gastric cancer cells
.
Br J Cancer
.
2018
. ;
118
(
6
):
867
-
877
.
61.
Paggetti
J
,
Haderk
F
,
Seiffert
M
, et al
.
Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts
.
Blood
.
2015
. ;
126
(
9
):
1106
-
1117
.
62.
Umezu
T
,
Tadokoro
H
,
Azuma
K
,
Yoshizawa
S
,
Ohyashiki
K
,
Ohyashiki
JH
.
Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1
.
Blood
.
2014
. ;
124
(
25
):
3748
-
3757
.
63.
Wieckowski
EU
,
Visus
C
,
Szajnik
M
,
Szczepanski
MJ
,
Storkus
WJ
,
Whiteside
TL
.
Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes
.
J Immunol
.
2009
. ;
183
(
6
):
3720
-
3730
.
64.
Szajnik
M
,
Czystowska
M
,
Szczepanski
MJ
,
Mandapathil
M
,
Whiteside
TL
.
Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory T cells (Treg)
.
PLoS One
.
2010
. ;
5
(
7
):
e11469
.
65.
Vences-Catalan
F
,
Rajapaksa
R
,
Srivastava
MK
, et al
.
Tetraspanin CD81 promotes tumor growth and metastasis by modulating the functions of T regulatory and myeloid-derived suppressor cells
.
Cancer Res
.
2015
. ;
75
(
21
):
4517
-
4526
.
66.
Tazi
I
,
Nafil
H
,
Mahmal
L
.
Monoclonal antibodies in hematological malignancies: past, present and future
.
J Cancer Res Ther
.
2011
. ;
7
(
4
):
399
-
407
.
67.
Vences-Catalan
F
,
Kuo
CC
,
Rajapaksa
R
, et al
.
CD81 is a novel immunotherapeutic target for B cell lymphoma
.
J Exp Med
.
2019
. ;
216
(
7
):
1497
-
1508
.
68.
Vences-Catalan
F
,
Rajapaksa
R
,
Kuo
CC
, et al
.
Targeting the tetraspanin CD81 reduces cancer invasion and metastasis
.
Proc Natl Acad Sci U S A
.
2021
. ;
118
(
24
):
e2018961118
.
69.
Nishioka
C
,
Ikezoe
T
,
Yokoyama
A
.
Blockade of CD82 by a monoclonal antibody potentiates anti-leukemia effects of AraC in vivo
.
Cancer Med
.
2015
. ;
4
(
9
):
1426
-
1431
.
70.
Heider
KH
,
Kiefer
K
,
Zenz
T
, et al
.
A novel Fc-engineered monoclonal antibody to CD37 with enhanced ADCC and high proapoptotic activity for treatment of B-cell malignancies
.
Blood
.
2011
. ;
118
(
15
):
4159
-
4168
.
71.
Algate
P
,
Liu
Z
,
Yuan
S
, et al
.
Abstract 2902: development of PSB202, a bifunctional antibody pair that target CD20 and CD37, for the treatment of B-cell malignancies
.
Cancer Res
.
2022
. ;
82
(
12 suppl
). 2902-2902.
72.
Betrian
S
,
Ysebaert
L
,
Heider
KH
,
Delord
JP
,
Fournié
JJ
,
Quillet-Mary
A
.
Idelalisib improves CD37 antibody BI 836826 cytotoxicity against chemo-resistant /relapse-initiating CLL cells: a rationale for combination treatment
.
Blood Cancer J
.
2016
. ;
6
(
11
):
e496
.
73.
Krause
G
,
Baki
I
,
Kerwien
S
, et al
.
Cytotoxicity of the CD37 antibody BI 836826 against chronic lymphocytic leukaemia cells in combination with chemotherapeutic agents or PI3K inhibitors
.
Br J Haematol
.
2016
. ;
173
(
5
):
791
-
794
.
74.
Geary
SM
,
Cambareri
AC
,
Sincock
PM
,
Fitter
S
,
Ashman
LK
.
Differential tissue expression of epitopes of the tetraspanin CD151 recognised by monoclonal antibodies
.
Tissue Antigens
.
2001
. ;
58
(
3
):
141
-
153
.
75.
Gutierrez-Lopez
MD
,
Ovalle
S
,
Yanez-Mo
M
, et al
.
A functionally relevant conformational epitope on the CD9 tetraspanin depends on the association with activated beta1 integrin
.
J Biol Chem
.
2003
. ;
278
(
1
):
208
-
218
.
76.
Jin
Y
,
Tachibana
I
,
Takeda
Y
, et al
.
Statins decrease lung inflammation in mice by upregulating tetraspanin CD9 in macrophages
.
PLoS One
.
2013
. ;
8
(
9
):
e73706
.
77.
Iwasaki
T
,
Takeda
Y
,
Maruyama
K
, et al
.
Deletion of tetraspanin CD9 diminishes lymphangiogenesis in vivo and in vitro
.
J Biol Chem
.
2013
. ;
288
(
4
):
2118
-
2131
.
78.
Knobeloch
KP
,
Wright
MD
,
Ochsenbein
AF
, et al
.
Targeted inactivation of the tetraspanin CD37 impairs T-cell-dependent B-cell response under suboptimal costimulatory conditions
.
Mol Cell Biol
.
2000
. ;
20
(
15
):
5363
-
5369
.
79.
van Spriel
AB
,
Puls
KL
,
Sofi
M
, et al
.
A regulatory role for CD37 in T cell proliferation
.
J Immunol
.
2004
. ;
172
(
5
):
2953
-
2961
.
80.
Sheng
KC
,
van Spriel
AB
,
Gartlan
KH
, et al
.
Tetraspanins CD37 and CD151 differentially regulate Ag presentation and T-cell co-stimulation by DC
.
Eur J Immunol
.
2009
. ;
39
(
1
):
50
-
55
.
81.
Wee
JL
,
Schulze
KE
,
Jones
EL
, et al
.
Tetraspanin CD37 regulates beta2 integrin-mediated adhesion and migration in neutrophils
.
J Immunol
.
2015
. ;
195
(
12
):
5770
-
5779
.
82.
Doyle
EL
,
Ridger
V
,
Ferraro
F
,
Turmaine
M
,
Saftig
P
,
Cutler
DF
.
CD63 is an essential cofactor to leukocyte recruitment by endothelial P-selectin
.
Blood
.
2011
. ;
118
(
15
):
4265
-
4273
.
83.
Tsitsikov
EN
,
Gutierrez-Ramos
JC
,
Geha
RS
.
Impaired CD19 expression and signaling, enhanced antibody response to type II T independent antigen and reduction of B-1 cells in CD81-deficient mice
.
Proc Natl Acad Sci U S A
.
1997
. ;
94
(
20
):
10844
-
10849
.
84.
Maecker
HT
,
Do
MS
,
Levy
S
.
CD81 on B cells promotes interleukin 4 secretion and antibody production during T helper type 2 immune responses
.
Proc Natl Acad Sci U S A
.
1998
. ;
95
(
5
):
2458
-
2462
.
85.
Lau
LM
,
Wee
JL
,
Wright
MD
, et al
.
The tetraspanin superfamily member CD151 regulates outside-in integrin alphaIIbbeta3 signaling and platelet function
.
Blood
.
2004
. ;
104
(
8
):
2368
-
2375
.
86.
Wright
MD
,
Geary
SM
,
Fitter
S
, et al
.
Characterization of mice lacking the tetraspanin superfamily member CD151
.
Mol Cell Biol
.
2004
. ;
24
(
13
):
5978
-
5988
.
87.
Higginbottom
A
,
Takahashi
Y
,
Bolling
L
, et al
.
Structural requirements for the inhibitory action of the CD9 large extracellular domain in sperm/oocyte binding and fusion
.
Biochem Biophys Res Commun
.
2003
. ;
311
(
1
):
208
-
214
.
88.
Barreiro
O
,
Yanez-Mo
M
,
Sala-Valdes
M
, et al
.
Endothelial tetraspanin microdomains regulate leukocyte firm adhesion during extravasation
.
Blood
.
2005
. ;
105
(
7
):
2852
-
2861
.
89.
Hemler
ME
.
Targeting of tetraspanin proteins--potential benefits and strategies
.
Nat Rev Drug Discov
.
2008
. ;
7
(
9
):
747
-
758
.
90.
Sharma
C
,
Yang
XH
,
Hemler
ME
.
DHHC2 affects palmitoylation, stability, and functions of tetraspanins CD9 and CD151
.
Mol Biol Cell
.
2008
. ;
19
(
8
):
3415
-
3425
.
91.
Latysheva
N
,
Muratov
G
,
Rajesh
S
, et al
.
Syntenin-1 is a new component of tetraspanin-enriched microdomains: mechanisms and consequences of the interaction of syntenin-1 with CD63
.
Mol Cell Biol
.
2006
. ;
26
(
20
):
7707
-
7718
.
92.
Dev
KK
.
Making protein interactions druggable: targeting PDZ domains
.
Nat Rev Drug Discov
.
2004
. ;
3
(
12
):
1047
-
1056
.
93.
Takeda
T
,
Hattori
N
,
Tokuhara
T
,
Nishimura
Y
,
Yokoyama
M
,
Miyake
M
.
Adenoviral transduction of MRP-1/CD9 and KAI1/CD82 inhibits lymph node metastasis in orthotopic lung cancer model
.
Cancer Res
.
2007
. ;
67
(
4
):
1744
-
1749
.
94.
Tsai
YC
,
Mendoza
A
,
Mariano
JM
, et al
.
The ubiquitin ligase gp78 promotes sarcoma metastasis by targeting KAI1 for degradation
.
Nat Med
.
2007
. ;
13
(
12
):
1504
-
1509
.
95.
Drucker
L
,
Tohami
T
,
Tartakover-Matalon
S
, et al
.
Promoter hypermethylation of tetraspanin members contributes to their silencing in myeloma cell lines
.
Carcinogenesis
.
2006
. ;
27
(
2
):
197
-
204
.
96.
Yu
J
,
Lee
CY
,
Changou
CA
,
Cedano-Prieto
DM
,
Takada
YK
,
Takada
Y
.
The CD9, CD81, and CD151 EC2 domains bind to the classical RGD-binding site of integrin alphavbeta3
.
Biochem J
.
2017
. ;
474
(
4
):
589
-
596
.