Anti-CD47 antibodies function primarily by blocking the CD47-SIRPα interaction, which normally serves as a "don't-eat-me" signal. CD47 is expressed on the surface of cells, including cancer cells, and inhibits phagocytosis by engaging the SIRPα receptor on phagocytes. When anti-CD47 antibodies bind to CD47, they disrupt this inhibitory signal, enabling increased phagocytosis of cancer cells by macrophages .
The mechanism involves multiple components:
Blockade of CD47-SIRPα interaction
Enhancement of phagocyte recognition of cancer cells
Facilitation of Fc receptor-dependent phagocytosis
Notably, experiments have confirmed that the Fc domain of anti-CD47 antibodies is essential for inducing phagocytosis. Blocking the Fc domain with anti-Fc F(ab')2 fragments or using F(ab')2 fragments derived from anti-CD47 antibodies prevents phagocytosis, demonstrating that Fc receptors are necessary for anti-CD47-driven phagocytosis .
CD47 has emerged as a promising therapeutic target because it is overexpressed in various malignancies, particularly hematological cancers like acute myeloid leukemia (AML) and non-Hodgkin's lymphoma (NHL). This overexpression helps cancer cells evade immune surveillance by inhibiting phagocytosis .
Research has demonstrated several key advantages of targeting CD47:
Widespread overexpression across multiple cancer types
Critical role in immune evasion through the macrophage checkpoint
Synergistic effects when combined with other therapies
Demonstrated efficacy in preclinical models leading to complete eradication of human AML in patient-derived xenografts
Pre-clinical studies have shown that humanized anti-CD47 antibodies such as Hu5F9-G4 can eliminate AML in vivo and lead to long-term disease-free survival in xenograft models, confirming CD47's value as a therapeutic target .
Distinguishing target-specific effects from off-target effects requires careful experimental design and controls. Researchers employ several methodological approaches:
Cell-specific pre-incubation experiments: Studies have shown that pre-incubating lymphoblasts with anti-CD47 antibodies followed by washing increases their phagocytosis three-fold, while pre-incubating macrophages has no effect. This confirms the effect is mediated through binding to the target cells rather than through effects on macrophages .
Receptor agonist controls: Researchers use CD47 receptor agonists like thrombospondin-1 or the peptide 4N1K to determine whether the effect is due to CD47 signaling activation. In controlled experiments, these agonists alone did not induce phagocytosis, suggesting the antibody effect is not simply due to CD47 activation .
Domain-specific blocking experiments: Blocking the Fc domain of anti-CD47 antibodies or using F(ab')2 fragments prevents phagocytosis, confirming that the effect depends on Fc receptor engagement rather than just CD47 blockade .
These methodologies help researchers separate direct effects on CD47-expressing target cells from potential off-target activities or non-specific immune activation.
Developing humanized anti-CD47 antibodies requires balancing efficacy, specificity, and safety through several technical considerations:
Antibody scaffold selection: IgG4 scaffolds are often preferred over IgG1 scaffolds for anti-CD47 antibodies because they minimize Fc-dependent effector functions like antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). This reduces potential damage to normal CD47-expressing cells .
Binding affinity optimization: Humanized antibodies like Hu5F9-G4 are designed with specific binding properties. For example, Hu5F9-G4 binds monomeric human CD47 with an 8 nM affinity, providing sufficient target engagement while minimizing potential toxicity .
Complementarity determining region (CDR) grafting: The humanization process involves grafting CDRs from murine antibodies onto human antibody frameworks. This requires careful selection of framework regions to maintain binding specificity and affinity .
Toxicokinetic profiling: Safety assessment in non-human primates is essential to determine safe dosing regimens that can achieve potentially therapeutic serum levels without unacceptable toxicity .
Combination strategy development: Due to the ubiquitous expression of CD47 on normal cells, single-agent therapy often has limited efficacy. Developing combination strategies (e.g., with rituximab for NHL or azacitidine for AML) is crucial for enhancing therapeutic outcomes .
These considerations are reflected in the development of antibodies like Hu5F9-G4, which progressed to clinical trials based on careful optimization of these parameters .
Differentiating phagocytosis of live versus dead cells is crucial for understanding the mechanism of anti-CD47 antibody-mediated cancer cell clearance. Researchers employ several methodological approaches:
Time-course analysis of cell viability: Studies have shown that anti-CD47 antibodies induce a rapid loss (within hours) of lymphoblasts in co-cultures with macrophages, without affecting lymphoblast viability in the absence of macrophages. This temporal relationship suggests phagocytosis of otherwise viable cells rather than antibody-induced cell death followed by phagocytosis .
Live cell imaging: Direct visualization of phagocytosis using time-lapse microscopy allows researchers to observe cancer cells being engulfed while still metabolically active and with intact membranes .
Vital dye exclusion: Combining phagocytosis assays with vital dyes that are excluded from live cells but enter dead cells helps determine whether cells are viable at the time of phagocytosis .
Inhibition of phagocytosis: Blocking phagocytosis using cytochalasin D or other phagocytosis inhibitors prevents cell death, confirming that death occurs as a result of phagocytosis (phagoptosis) rather than preceding it .
Through these approaches, researchers have established that anti-CD47 antibodies induce phagocytosis of otherwise live cancer cells, and that the cell death occurs as a direct result of this phagocytosis—a process termed "phagoptosis" .
Evaluating synergy between anti-CD47 antibodies and other therapeutics requires robust experimental approaches:
In vitro phagocytosis assays: Researchers assess the phagocytic index (percentage of macrophages that have engulfed cancer cells) when cancer cells are treated with anti-CD47 antibodies alone or in combination with other agents. This allows quantification of enhanced phagocytosis beyond additive effects .
Patient-derived xenograft models: Studies have demonstrated that while humanized anti-CD47 antibodies like Hu5F9-G4 can eliminate AML in xenograft models as monotherapy, they synergize with rituximab to eliminate NHL engraftment and cure xenografted mice, providing compelling evidence of in vivo synergy .
Mechanistic studies of combinatorial effects:
Isobologram analysis: This mathematical approach determines whether drug combinations produce effects greater than the sum of their individual effects, allowing quantitative assessment of synergy versus additivity .
Several clinical trials are now evaluating these synergistic combinations, including anti-CD47 antibodies with azacitidine for AML/MDS, and with rituximab or tislelizumab for lymphomas, based on preclinical evidence of synergy .
Optimal assessment of anti-CD47 antibody-mediated phagocytosis requires careful experimental design:
Cell labeling approach:
Target cells (e.g., cancer cells) should be fluorescently labeled to allow quantitative measurement of phagocytosis
Common fluorophores include CFSE, CellTracker dyes, or pH-sensitive dyes that change fluorescence upon internalization into acidic phagosomes
Macrophage preparation:
Both cell lines (e.g., U937) and primary macrophages derived from peripheral blood monocytes should be used
Macrophage polarization state (M1 vs. M2) can significantly affect phagocytic capacity and should be characterized and reported
Co-culture conditions:
Quantification methods:
Flow cytometry for high-throughput assessment (percentage of macrophages containing fluorescent target cells)
Confocal microscopy for visual confirmation of internalization versus surface binding
Live cell imaging for real-time assessment of phagocytosis kinetics
Essential controls:
These protocols have revealed that anti-CD47 antibodies can increase phagocytosis by macrophages of various B-cell lines (697, Ramos, DG-75) by several fold compared to controls, with significant depletion (approximately 75%) of target cells after 6 hours of co-culture .
Addressing CD47 expression on normal cells presents significant challenges for in vivo studies. Researchers should implement the following strategies:
Antibody engineering approaches:
Use of IgG4 scaffolds rather than IgG1 to minimize Fc-dependent effector functions against normal cells
Development of bispecific antibodies that target CD47 and tumor-specific antigens (e.g., CD19, CD20) to enhance tumor selectivity
"Imbalanced" design with decreased binding affinity to CD47 and increased affinity to tumor cell surface proteins to enhance specificity
Dosing strategy considerations:
Toxicity monitoring:
Regular assessment of complete blood counts to monitor for anemia and thrombocytopenia
Spleen size monitoring for potential sequestration
Assessment of liver enzymes and other parameters to detect off-target effects
Novel delivery systems:
Nanoparticle-based delivery to enhance tumor-specific targeting
Iron oxide magnetic nanoparticles as carriers of anti-CD47 antibodies can increase delivery to cancer cells while reducing systemic exposure
Controlled release formulations that maintain therapeutic levels while minimizing peak concentrations that might affect normal tissues
These approaches have enabled successful in vivo studies with anti-CD47 antibodies, leading to complete eradication of human AML and long-term disease-free survival in patient-derived xenograft models while managing toxicity to normal tissues .
Development of bispecific antibodies targeting CD47 and tumor-specific antigens requires attention to several technical parameters:
Target selection and validation:
Binding domain optimization:
Affinity balancing between the two binding arms is critical
Higher affinity for the tumor-specific antigen compared to CD47 improves specificity
For example, IMM0306 (targeting CD47 and CD20) was engineered with higher affinity for CD20, resulting in preferential binding to malignant B cells and more effective anti-lymphoma activity
Format selection:
Various bispecific formats (e.g., dual-variable domain, diabody, tandem scFv) have different pharmacokinetic profiles
Fc inclusion affects half-life and potential effector functions
Molecular size impacts tumor penetration and biodistribution
Functional evaluation:
Assessment of both CD47 blockade and engagement of the tumor-specific target
Confirmation that dual targeting enhances phagocytosis beyond single-target approaches
Verification that normal cells expressing only CD47 are spared
Several bispecific antibodies are in clinical development, including TG-1801 (CD47/CD19) which, in combination with ublituximab (anti-CD20), achieved a 44% objective response rate in relapsed/refractory B-cell lymphoma patients, including complete responses .
Novel drug delivery systems show significant promise for improving anti-CD47 antibody efficacy while reducing toxicity:
Nanoparticle-based delivery platforms:
Multifunctionalized iron oxide magnetic nanoparticles can serve as carriers for anti-CD47 antibodies, preserving their targeting activity while increasing delivery to cancer cells and accelerating cancer cell apoptosis
Nanoparticles allow controlled release of anti-CD47 antibodies, maintaining therapeutic levels at tumor sites while minimizing systemic exposure
Tumor microenvironment-responsive systems:
pH-sensitive nanocarriers can release anti-CD47 antibodies preferentially in the acidic tumor microenvironment
Enzyme-cleavable linkers activated by tumor-associated proteases can enable site-specific antibody release
Combination delivery systems:
Co-delivery of anti-CD47 antibodies with agents that increase prophagocytic signals (e.g., calreticulin inducers)
Nanoparticles carrying both anti-CD47 antibodies and chemotherapeutics for synergistic effects
Physical targeting methods:
Magnetic guidance for iron oxide nanoparticles carrying anti-CD47 antibodies
Photodynamic therapy sensitizers co-delivered with anti-CD47 antibodies for site-specific activation
These approaches address the fundamental challenge of CD47-targeted therapies: delivering effective treatment to cancer cells while minimizing effects on normal CD47-expressing cells. In mouse models, nanoparticles loaded with anti-CD47 antibodies have achieved significant antitumor effects by continuously releasing antibodies to block the CD47-SIRPα axis .
Several strategies are being developed to address potential resistance mechanisms to anti-CD47 antibody therapy:
Targeting alternative "don't eat me" signals:
Some cancer cells may upregulate alternative inhibitory signals beyond CD47
Combination approaches targeting multiple inhibitory pathways simultaneously may prevent resistance development
Enhancing "eat me" signals:
Leveraging adaptive immunity:
Novel bispecific formats:
Phagocyte-activating strategies:
Combination with macrophage-polarizing agents to enhance phagocytic capacity
Use of GM-CSF or other cytokines to increase numbers and activity of phagocytes
These approaches recognize that cancer cells employ multiple immune evasion strategies and that combination approaches targeting complementary pathways are likely to be most effective in preventing or overcoming resistance.
The tumor microenvironment (TME) significantly impacts anti-CD47 antibody efficacy through several mechanisms:
Macrophage polarization state:
M2-polarized (immunosuppressive) macrophages predominate in many tumors and may have reduced phagocytic capacity
Repolarizing macrophages toward an M1 phenotype using agents like TLR agonists can enhance anti-CD47 antibody efficacy
Competitive inhibition by soluble factors:
Hypoxia and metabolic factors:
Hypoxic conditions can alter macrophage function and phagocytic capacity
Tumor cell metabolism produces factors that may inhibit macrophage activity
Extracellular matrix barriers:
Dense extracellular matrix may limit antibody penetration and macrophage mobility
Matrix-degrading enzymes or agents that normalize tumor vasculature may improve delivery
Potential strategies to address these challenges include:
Combination with macrophage-repolarizing agents
Use of matrix-modifying enzymes to enhance antibody penetration
Development of bispecific antibodies that can recruit and activate macrophages while blocking CD47
Combination with vascular normalization strategies to improve tissue penetration
Understanding and manipulating the TME may be critical for maximizing the efficacy of anti-CD47 antibodies across different tumor types and microenvironmental contexts.