Fact-checked by Grok 2 weeks ago

Complement-dependent cytotoxicity

Complement-dependent cytotoxicity (CDC) is an effector mechanism of the in which antibodies, primarily IgG or IgM, bind to antigens on the surface of target cells, activating the to form the membrane attack complex (), which inserts into the and causes . This process engages both innate and adaptive immunity, serving as a key defense against pathogens, infected cells, and abnormal cells such as tumors. The mechanism involves the Fc region of bound antibodies recruiting C1q to initiate the complement cascade, leading to proteolytic cleavages that deposit C3b, generate anaphylatoxins (C3a, C5a), and assemble the MAC (C5b-9) for cell lysis. Efficiency varies with antibody isotype (e.g., IgG1 and IgM effective; IgG4 inactive), antigen density, and regulators like complement factor H. In therapeutic contexts, CDC mediates action for monoclonal antibodies (mAbs) in and autoimmune diseases, such as anti-CD20 agents rituximab and targeting B-cell malignancies. Fc engineering strategies to enhance hexamerization or counter inhibitors have yielded several such antibodies in clinical trials. In vivo challenges, including complement regulators and tumor penetration, drive research into combinations.

Biological Basis

Definition

Complement-dependent cytotoxicity (CDC) is an immune-mediated process in which antibodies bound to antigens on the surface of target cells activate the , resulting in the disruption and of the target cell membrane. This mechanism serves as a key effector function of , enabling the elimination of pathogens, infected cells, or abnormal cells such as tumors. CDC requires the formation of antibody-antigen complexes on the cell surface to initiate complement activation, distinguishing it from other antibody-dependent killing pathways. The process primarily involves IgM and certain subclasses of IgG antibodies, with IgG1 and IgG3 being the most effective at recruiting complement due to their ability to bind C1q, the initiating protein of the classical pathway. IgM, often pentameric or hexameric in structure, is particularly potent in activating complement even at low densities, while IgG subclasses facilitate multimeric binding that enhances C1q interaction. Complement proteins, starting with C1q, are essential components that recognize and bind to the Fc regions of these surface-bound antibodies, setting the stage for downstream cascade events. As part of the , the provides a rapid, antibody-assisted defense mechanism that bridges innate and adaptive immunity, though CDC specifically depends on pre-existing or induced antibodies for targeting. This prerequisite of antibody-antigen complexes on the target cell surface ensures specificity, preventing indiscriminate activation of the complement cascade in the absence of immune recognition.

Role in Innate Immunity

Complement-dependent cytotoxicity (CDC) serves as a critical bridge between the adaptive and innate immune systems, where antibodies produced by B cells from the adaptive response bind to pathogens or infected cells, thereby activating the complement cascade to initiate innate effector functions. This integration enhances pathogen clearance by leveraging the rapid, non-specific responses of the innate system to amplify antibody-mediated targeting. In innate immunity, CDC plays a key defensive role against and viruses by forming the membrane attack complex (MAC) on their surfaces, leading to and destruction of these pathogens or virus-infected cells. For instance, it contributes to the elimination of enveloped viruses through neutralization and the killing of infected cells, while also aiding in bacterial clearance by disrupting microbial membranes. Additionally, complement activation during CDC generates fragments such as C3b for opsonization, marking targets for by innate immune cells like macrophages and neutrophils, and anaphylatoxins C3a and C5a that promote by recruiting and activating these cells to the site of . The underlying CDC has evolved in vertebrates as an ancient component of innate immunity, providing a first line of defense against invading microbes across species. To prevent unintended damage to host tissues, physiological regulation occurs through membrane-bound inhibitors such as (decay-accelerating factor), which disrupts C3 convertases, and , which inhibits assembly on healthy cells. This tight control ensures that CDC's lytic potential is directed primarily toward foreign or altered self-cells while minimizing autoinflammatory effects.

Mechanism of Action

Antibody Binding and Complement Activation

Complement-dependent cytotoxicity (CDC) begins with the binding of to specific antigens on the surface of target cells, primarily through the Fab regions of (IgG) subclasses IgG1 and IgG3 or (IgM). This interaction forms immune complexes on the , positioning the regions for subsequent complement engagement. The is initiated when the C1q component of the C1 complex binds to the regions of these surface-bound antibodies. For IgG, effective C1q binding requires at least two closely spaced regions, often achieved through the formation of IgG hexamers that cluster multiple domains for high-avidity interaction with C1q's globular heads. In contrast, a single pentameric IgM molecule suffices due to its multivalent structure, which presents multiple regions simultaneously. Upon binding, C1q undergoes a conformational change that triggers autoactivation of C1r, which in turn activates the C1s within the C1 complex. Activated C1s then cleaves complement component into C4a and C4b, with C4b covalently attaching to the target cell surface via thioester bonds. Subsequently, C1s cleaves into C2a and C2b; the C4b-C2a complex forms the (C4b2a), which proteolytically cleaves into C3a and C3b. C3b deposition amplifies the cascade by facilitating further convertase assembly and opsonization, marking the target for immune clearance. The efficiency of this process is heavily influenced by the density of antigens on the target cell and the of antibody binding. High antigen density promotes IgG hexamerization and optimal Fc clustering, enhancing C1q recruitment, whereas low density may require multiple IgG molecules (typically six or more in a hexameric arrangement) to achieve sufficient for activation. Suboptimal spacing or insufficient antibody occupancy can limit cascade initiation, underscoring the importance of epitope proximity and antibody valency in CDC potency.

Formation of Membrane Attack Complex

The terminal phase of complement activation in complement-dependent cytotoxicity is initiated by , a complex formed after C3b deposition on the antibody-opsonized target cell, which cleaves the complement protein into the anaphylatoxin C5a and the fragment C5b. promotes inflammation by recruiting immune cells, while C5b binds to the and serves as the nucleating center for the terminal complement pathway. Assembly of the membrane attack complex (), also known as C5b-9, proceeds through sequential binding of additional complement proteins to C5b. C5b first associates with to form the stable C5b-6 intermediate, followed by the binding of C7, which induces a conformational change that anchors the complex to the via hydrophobic interactions. C8 then attaches to the C5b-6-7 complex, with its α subunit facilitating initial membrane penetration, and finally, multiple C9 molecules (typically 12-18) polymerize around the structure, forming a cylindrical, pore-like transmembrane channel approximately 100 in inner and up to 240 in outer . This oligomerization creates a stable, β-barrel-shaped scaffold that completes the . The insertion of the into the target disrupts its integrity by forming a hydrophilic that permits uncontrolled influx of water and ions, leading to osmotic swelling, colloid osmotic , and ultimately . The 's size allows passage of molecules up to 10 kDa, ensuring effective of susceptible cells such as or nucleated cells lacking protective regulators. To prevent inadvertent damage to host cells, formation is tightly regulated by soluble and membrane-bound inhibitors. , also known as S-protein, binds to the C5b-7 intermediate in the fluid phase, stabilizing it and inhibiting its insertion into membranes or subsequent recruitment of C8 and C9. Membrane-bound regulators like further block C9 polymerization on host cells, limiting assembly and formation.

Assay Methods

Principles of CDC Assays

Complement-dependent cytotoxicity (CDC) assays are tests designed to evaluate the ability of antibodies in to trigger complement-mediated of target s expressing specific antigens. The exploits the biological where antibodies bind to surface antigens, activating the to form the membrane attack complex (MAC), which perforates the and causes . Target s, such as lymphocytes or tumor cell lines, are typically labeled with a , radioisotope, or other marker prior to incubation with patient containing potential antibodies, followed by the addition of exogenous complement, often sourced from rabbit to ensure consistent activity. This setup allows for the quantification of antibody-dependent complement activation without relying on the subject's endogenous complement levels. The foundational CDC assay, known as the microlymphocytotoxicity test, originated in the 1960s as a tool for detecting cytotoxic antibodies, particularly in the context of histocompatibility testing. Developed by Terasaki and McClelland in 1964, it involved miniaturizing the assay into microdroplets using Terasaki plates, enabling high-throughput screening of serum samples against panels of target cells. Over time, the assay evolved to include more quantitative methods, with the ⁵¹Cr-release assay emerging as a gold standard for precise measurement of cell lysis in research settings; in this variant, target cells are pre-loaded with radioactive chromium-51, and the percentage of isotope released into the supernatant correlates directly with the extent of cytotoxicity. Incubation conditions are standardized, typically involving 30-60 minutes at room temperature or 37°C for antibody binding, followed by 1-4 hours with complement to allow MAC formation and lysis. Cytotoxicity in CDC assays is quantified by indicators of cell death, such as percentage determined via exclusion (where viable cells exclude the dye while dead cells take it up, scored microscopically on a scale of 1-8) or release of intracellular enzymes like (LDH) in non-radioactive endpoints. Flow cytometry-based methods represent a modern endpoint variant, using fluorescent dyes (e.g., propidium iodide) to distinguish live from dead cells after staining, offering higher sensitivity and objectivity compared to traditional visual scoring. To ensure reliability, assays incorporate positive controls (e.g., serum with known high-titer antibodies plus complement to achieve >80% ) and negative controls (e.g., serum alone or complement alone, yielding <10% ), along with complement titration to optimize activity and account for batch variability. Standardization follows guidelines from bodies like the American Society for Histocompatibility and Immunogenetics (), emphasizing consistent cell concentrations (around 1-3 × 10⁶ cells/mL) and reagent quality to minimize false positives or negatives.

Detection Techniques

Complement-dependent cytotoxicity (CDC) assays traditionally rely on the 51Cr-release method to quantify , where target cells are pre-labeled with radioactive , and the release of the into the supernatant following complement-mediated damage serves as a direct measure of . This technique offers high sensitivity for detecting low levels of , making it suitable for precise quantification in research settings, though it requires handling radioactive materials, which poses safety and disposal challenges. Dye exclusion assays represent another classical approach, utilizing vital dyes such as or to distinguish viable cells (which exclude the dye) from dead cells (which take up the dye due to compromised membranes). In standard CDC protocols for anti-HLA antibody detection, cells are incubated with and complement, followed by dye addition and microscopic evaluation, with results scored on a scale of 1-8 based on the percentage of stained (lysed) cells, where scores ≥2 indicate positive . These methods are simple and cost-effective but are subjective and labor-intensive, limiting throughput. Modern detection techniques have shifted toward non-radioactive, high-throughput options, including with fluorescent labels like propidium iodide (PI), which intercalates into the DNA of cells with permeabilized membranes to identify lysed populations. To differentiate true complement-induced from , dual staining with annexin V (which binds exposed on apoptotic cells) and PI is employed; PI-positive, annexin V-negative cells indicate necrotic , while annexin V-positive, PI-negative cells signify early . provides advantages in multiplexing, allowing simultaneous assessment of cell subsets and binding, with greater objectivity and sensitivity compared to traditional microscopy-based scoring. However, it requires specialized equipment and can be affected by non-specific . Luminescence-based assays offer an alternative for quantifying through the detection of intracellular contents released upon , such as ATP, using luciferase-mediated for a sensitive, readout. These methods are advantageous for their simplicity, avoidance of , and compatibility with high-throughput formats like 96-well plates, though they may suffer from by serum components in complement-containing reactions. Across these techniques, specific lysis is calculated to normalize for background cell death using the formula: \% \text{ specific lysis} = \frac{\text{experimental release} - \text{spontaneous release}}{\text{maximum release} - \text{spontaneous release}} \times 100 where experimental release reflects in the presence of and complement, spontaneous release is from untreated cells, and maximum release is from detergent-lysed cells. This metric establishes the - and complement-dependent component of , enabling comparison across assays despite varying detection modalities.

Clinical Applications

Therapeutic Monoclonal Antibodies

Therapeutic monoclonal antibodies (mAbs) harness complement-dependent cytotoxicity (CDC) as a key mechanism to eliminate target cells in cancer and autoimmune diseases, marking a significant advancement since the first CDC-active mAb approvals in the , such as rituximab in for relapsed or refractory low-grade or follicular B-cell non-Hodgkin's . Rituximab, an anti- IgG1 mAb, binds to on malignant B cells, initiating the by recruiting C1q to form the membrane attack complex () that lyses target cells. In B-cell , this CDC activity contributes substantially to tumor clearance, with preclinical models demonstrating that complement depletion reduces rituximab's antitumor effects. Another example is trastuzumab, an anti-HER2 IgG1 mAb used in HER2-overexpressing , where CDC provides a partial but supportive role alongside (), though its potency is often limited by overexpression of complement regulatory proteins like CD55 and on tumor cells. To optimize CDC, mAbs are engineered through Fc region modifications that enhance C1q binding and complement activation. Glycosylation alterations in the Fc domain, such as introducing specific substitutions in the CH2 region (e.g., S267E/H268F/S324T), increase C1q up to 23-fold, boosting CDC against CD20-positive tumor s in vitro. Afucosylation of the Fc-linked N297 primarily augments ADCC by improving FcγRIIIa binding but also synergizes with CDC by facilitating denser clustering on cell surfaces, enhancing overall effector in therapies. These engineered variants, like (an afucosylated anti-CD20 mAb), demonstrate superior CDC in preclinical studies compared to rituximab, particularly in complement-limited environments. Clinical evidence underscores CDC's importance, as seen in studies where rituximab efficacy is diminished in patients with complement deficiencies, such as those with (CLL) exhibiting low C1q levels or acquired complement defects, leading to reduced CDC-mediated B-cell depletion. In complement-deficient models, rituximab's tumor clearance drops significantly, highlighting CDC's non-redundant role . Dosing regimens must account for complement activation to mitigate infusion-related reactions (IRRs), which correlate with rapid consumption and C5a generation post-infusion; slower initial dosing (e.g., 50 mg/hour ramp-up) reduces IRR incidence from 77% to 15% while preserving efficacy. CDC assays are routinely used in potency testing to ensure batch consistency for these mAbs.

HLA Typing and Crossmatching

In the context of solid organ transplantation, particularly kidney and heart procedures, the complement-dependent cytotoxicity (CDC) crossmatch serves as a critical diagnostic tool to detect preformed donor-specific anti-HLA antibodies in recipient serum, thereby assessing the risk of hyperacute rejection. The procedure involves mixing recipient serum with isolated donor lymphocytes, typically T cells for HLA class I detection and B cells for both class I and II, followed by the addition of rabbit complement; complement activation by donor-specific antibodies leads to cell lysis, visualized through dye exclusion or fluorescence microscopy. HLA typing is integrated with CDC assays through panel-reactive antibody (PRA) testing, which quantifies the percentage of reactivity against a panel of HLA-typed donor cells, indicating the recipient's sensitization level and likelihood of a positive crossmatch with potential donors. This approach, established as a historical standard since the 1960s following the development of the CDC method by Terasaki and colleagues, has been essential for prioritizing low-sensitization patients in kidney and heart transplant allocation. Interpretation of CDC crossmatch results relies on the extent of observed : a positive result, indicating complement-mediated , predicts a high of immediate graft failure, as demonstrated in seminal 1969 studies where 24 of 30 transplants across positive crossmatches failed rapidly compared to only 8 of 195 with negative results. Specific protocols emphasize separating T and B cells via magnetic bead isolation or rosetting to enhance specificity, with scoring based on the of dead cells—typically a cutoff of greater than 20% (or a score of 2 or higher on a 0-8 scale) denoting positivity. Over time, while CDC remains a for its functional assessment of complement-activating antibodies, it has evolved alongside more sensitive methods like , which detects lower-level antibodies to reduce false negatives in high-risk transplants.

Limitations and Considerations

Factors Affecting Efficacy

The efficacy of complement-dependent cytotoxicity (CDC) is modulated by several biological factors, including genetic deficiencies in complement components and the density of target antigens on cell surfaces. Individuals with hereditary deficiencies in early classical pathway components, such as or , exhibit significantly reduced CDC against antibody-opsonized targets, as these mutations impair the initiation of the cascade required for C1q binding and downstream activation. Similarly, deficiencies in terminal components like limit (MAC) formation, though engineered antibodies can partially overcome this by promoting hexamerization under complement-limiting conditions. Low target antigen density, as observed on (CLL) cells expressing reduced compared to B-cell cells, impairs CDC initiation by hindering sufficient antibody clustering for effective C1q recruitment, often resulting in minimal even with potent monoclonal antibodies like rituximab. Environmental variables further influence CDC in vivo, particularly variations in serum complement levels and the presence of endogenous inhibitors. Serum concentrations of complement proteins, such as and , fluctuate with age and sex; for instance, pathway activity is approximately 14% lower in females than males, while classical and pathway activities increase with age in healthy populations, by approximately 12-16% from young adulthood to older age. Diseases including autoimmune disorders and malignancies can deplete complement through consumption or altered production, reducing available components for CDC and thereby limiting therapeutic antibody efficacy in affected patients. Soluble inhibitors like (CR1) and (fH) block activation by accelerating decay of convertases or serving as cofactors for C3b inactivation; for example, elevated fH on tumor cells via extracellular vesicles can suppress CDC by over 50% in preclinical models, while CR1 neutralization has been shown to enhance antibody-mediated . In settings, procedural factors introduce variability that affects measured . The choice of complement is critical, as often yields 20-80% lower compared to complement due to species-specific differences in regulatory protein interactions, such as reduced inhibition by CD55 in systems, making complement preferable for clinically relevant assessments. Temperature and pH also impact cascade efficiency; at 31°C doubles antibody-initiated CDC compared to 37°C by enhancing insertion, whereas acidic conditions (pH 5.5-6.0), common in tumor microenvironments, inhibit all complement pathways by over 50% through impaired / deposition and convertase stability. Quantitative studies on antibody reveal that high- (K_D < 2 nM) can reduce by 60-70% relative to optimized lower- variants (K_D 20-80 nM), as excessive target engagement disrupts clustering needed for C1q , underscoring the need for tuning in therapeutic design.

Alternatives to CDC

Antibody-dependent cellular cytotoxicity (ADCC) serves as a primary alternative to complement-dependent cytotoxicity (CDC) in antibody-mediated immune responses, wherein cells and other effector cells recognize antibody-coated target cells through the low-affinity Fcγ receptor IIIa (FcγRIIIa, also known as ). This binding triggers the release of cytotoxic granules, including perforin and granzymes, leading to target cell without reliance on the complement cascade. Glycoengineering of therapeutic antibodies, such as the defucosylation in , enhances FcγRIIIa affinity, significantly boosting ADCC efficacy in clinical settings like treatment. Complement-independent phagocytosis represents another key mechanism, where C3b fragments generated during early complement activation opsonize pathogens or tumor cells for uptake by expressing complement receptors (e.g., CR1 and CR3), promoting lysosomal degradation without the need for membrane attack complex ()-induced . This process enhances clearance in tissues where complement proteins may be scarce or where lytic activity is undesirable, as seen in immune responses to encapsulated . Emerging therapeutic strategies further diversify options beyond CDC. Antibody-drug conjugates (ADCs) link monoclonal antibodies to potent cytotoxic payloads via chemical linkers, enabling targeted delivery and intracellular release of toxins like auristatins or maytansinoids, thereby bypassing complement activation entirely for direct cell killing. Similarly, chimeric antigen receptor T (CAR-T) cell therapies engineer patient-derived T cells to express synthetic receptors that recognize tumor and mediate perforin/granzyme-dependent , offering a cellular alternative independent of . In comparative terms, ADCC often predominates over CDC in solid tumors due to limited complement penetration into dense stromal environments, reducing formation efficacy while cell infiltration supports antibody-directed killing. This has driven a historical shift in development during the , from CDC-focused designs like rituximab to ADCC-optimized variants, reflecting improved clinical outcomes in non-hematologic malignancies.

References

  1. [1]
    Complement-Dependent Cytotoxicity (CDC) - Sino Biological
    Complement-dependent cytotoxicity (CDC) is initiated by C1q, the initiating component of the classical complement pathway.
  2. [2]
    Regulation of antibody-mediated complement-dependent ... - PMC
    Dec 12, 2019 · Complement-dependent cytotoxicity (CDC) is a potent effector mechanism, engaging both innate and adaptive immunity.Results · Reducing Igg Intrinsic... · Cell Binding By AntibodiesMissing: definition | Show results with:definition
  3. [3]
    Complement-dependent cytotoxicity (CDC) - ScienceDirect.com
    Complement Dependent Cytotoxicity (CDC) is a process where monoclonal antibodies activate the complement pathway, leading to the formation of the membrane ...
  4. [4]
  5. [5]
    Enhancement of complement-dependent cytotoxicity by linking ...
    Antibody-mediated complement-dependent cytotoxicity (CDC) on malignant cells is regulated by several complement control proteins, including the inhibitory ...
  6. [6]
    The complement system and innate immunity - Immunobiology - NCBI
    The complement system is one of the major mechanisms by which pathogen recognition is converted into an effective host defense against initial infection.2-5. Complement is a system... · 2-9. Hydrolysis of C3 causes...
  7. [7]
    Complement Dependent Cytotoxicity - an overview
    Complement dependent cytotoxicity (CDC) is defined as a mechanism through which antibodies, primarily IgG1 and IgG3, activate a cascade of ...
  8. [8]
    The Influence of Human IgG Subclass and Allotype on Complement ...
    At critically low antigen densities, IgM hexamers outcompete both IgM pentamers and IgG1 for human complement deposition and complement-dependent cytotoxicity.
  9. [9]
    Complement and its role in innate and adaptive immune responses
    Dec 15, 2009 · Complement is a functional bridge between innate and adaptive immune responses that allows an integrated host defense to pathogenic challenges.
  10. [10]
    Immune Defenses - Medical Microbiology - NCBI Bookshelf - NIH
    Antibody and complement acting together can inactivate certain viruses (in most cases, enveloped viruses). Antibody is most effective against virus in large ...
  11. [11]
    Viral Evasion of the Complement System and Its Importance for ...
    The complement system can directly neutralize virus particles through opsonisation (3), membrane attack complex (MAC) formation on the virion (4), MAC formation ...
  12. [12]
    Complement System Part II: Role in Immunity - PMC
    Anaphylatoxins C3a and C5a participate in inflammation by interacting and activating immune cells via C3aR and C5aR, respectively. C3a is implicated in the ...
  13. [13]
    The 'Ins and Outs' of complement-driven immune responses - PMC
    The complement system represents an evolutionary old and critical component of innate immunity where it forms the first line of defence against invading ...
  14. [14]
    The relevance of complement to virus biology - PMC - PubMed Central
    CD59 is a cell surface protein that prevents formation of the terminal membrane attack complex (MAC) involved in membrane disruption and cell lysis. RCA ...
  15. [15]
  16. [16]
  17. [17]
  18. [18]
  19. [19]
  20. [20]
  21. [21]
    Complement Membrane Attack Complex: New Roles, Mechanisms ...
    The complement membrane attack complex (MAC) is classically known as a cytolytic effector of innate and adaptive immunity that forms pores in the plasma ...
  22. [22]
    Structural basis of complement membrane attack complex formation
    Feb 4, 2016 · In response to complement activation, the membrane attack complex (MAC) assembles from fluid-phase proteins to form pores in lipid bilayers.
  23. [23]
  24. [24]
    a review of the complement membrane attack complex - PMC
    Jun 19, 2017 · The membrane attack complex (MAC) is an important innate immune effector of the complement terminal pathway that forms cytotoxic pores on the surface of ...
  25. [25]
    Complement‐Dependent Cytotoxicity (CDC) to Detect Anti‐HLA ...
    The method to detect anti‐HLA antibodies has historically been the complement‐dependent cytotoxicity (CDC) assay, described by Terasaki and McClelland in 1964 ...Assays · Cdc Assay · Pra Detected By Spi
  26. [26]
  27. [27]
  28. [28]
    Complement-dependent serum cytotoxicity of cancer patients ...
    The conditions for a 51Cr release assay on cell lines derived from human solid tumors were determined by using a rabbit antihuman antiserum with human AB ...
  29. [29]
    Potent complement-dependent cytotoxicity of tumor cells by target ...
    Jul 15, 2016 · ... complement-dependent cytotoxicity (CDC) as single agents. ... CDC activity was assessed by 51Cr release assays in the presence of human serum.
  30. [30]
    Real Time Assays for Quantifying Cytotoxicity with Single Cell ...
    A new live cell-based assay platform has been developed for the determination of complement dependent cytotoxicity (CDC), antibody dependent cellular ...
  31. [31]
    A non-radioactive complement-dependent cytotoxicity assay for anti ...
    The assay measures the relative number of viable cells based on the uptake and metabolism of the redox dye, Alamar blue. ... exclusion of Trypan blue by viable ...
  32. [32]
    FDA/PI flow cytometry assay of complement‐mediated cytotoxicity of ...
    Sep 28, 2004 · A flow cytometry complement-mediated cytotoxicity assay (FCCA) using fluorescein diacetate (FDA) and propidium iodide (PI) to measure antibody-dependent ...
  33. [33]
    Protocol for Apoptosis Assay by Flow Cytometry Using Annexin V ...
    The apoptosis assay uses Annexin V and propidium iodide staining, followed by flow cytometry. PI-negative, Annexin V-positive cells indicate apoptosis.
  34. [34]
    Evaluation of complement-dependent cytotoxicity using ATP ...
    The luminescence method coupled with complement protein analysis by flow cytometry encompasses all needed methods to evaluate antibody ability to trigger CDC.Missing: review principles
  35. [35]
    Evaluation of Complement-Dependent Cytotoxicity Using ATP ...
    The luminescence method coupled with complement protein analysis by flow cytometry encompasses all needed methods to evaluate antibody ability to trigger CDC.
  36. [36]
    In vitro mechanisms of action of rituximab on primary non-Hodgkin ...
    Feb 1, 2003 · The cytotoxicity of effector cells in the presence or absence of 2 μg/mL rituximab was measured in a standard 4-hour51Cr-release assay, as ...
  37. [37]
    Search Orphan Drug Designations and Approvals - FDA
    Marketing approved: 1, Generic Name: rituximab. Trade Name: Rituxan. Marketing Approval Date: 11/26/1997. Approved Labeled Indication: Treatment of patients ...
  38. [38]
    the first monoclonal antibody approved for the treatment of lymphoma
    Rituximab, a genetically engineered monoclonal chimeric antibody, targets the CD20 antigen expressed on B cells. It was approved by the US Food and Drug ...
  39. [39]
    Rituximab: mechanism of action - PMC - NIH
    Direct signaling, complement dependent cellular cytotoxicity and antibody dependent cellular cytotoxicity all appear to play a role in rituximab efficacy.
  40. [40]
    The role of complement in the mechanism of action of rituximab for B ...
    Sep 8, 2008 · Rituximab significantly improves treatment outcome in relapsed or refractory, low-grade or follicular B-cell non-Hodgkin's lymphoma (NHL).
  41. [41]
    Complement activation determines the therapeutic activity ... - PubMed
    A single injection of rituximab or the murine anti-CD20 Ab 1F5, given i.p. 1 day after the tumor, cured 100% of the animals. Indeed, at week 4 after tumor cell ...
  42. [42]
    CD55 and CD59 expression protects HER2-overexpressing breast ...
    It was hypothesized that CDC induced by trastuzumab in HER2-positive breast cancer may be limited due to overexpression of CD55 and CD59. The aim of the present ...
  43. [43]
    CD55 and CD59 expression protects HER2-overexpressing breast ...
    Jul 8, 2017 · ... breast cancer do not benefit from trastuzumab treatment, potentially due to the lack of complement-dependent cytotoxicity (CDC) activation.
  44. [44]
    Engineered Fc variant antibodies with enhanced ability to recruit ...
    ... complement- dependent cytotoxicity (CDC), antibody-dependent cellular ... Engineered antibodies of IgG1/IgG3 mixed isotype with enhanced cytotoxic activities.<|control11|><|separator|>
  45. [45]
    Engineered Fc variant antibodies with enhanced ability to ... - PubMed
    Variants enhanced the cytotoxic potency of an anti-CD20 antibody up to 23-fold against tumor cells in CDC assays, and demonstrated a correlated increase in C1q ...
  46. [46]
    Enhancing Fc‐mediated effector functions of monoclonal antibodies
    Sep 14, 2024 · In this review, we will outline the different approaches to enhance Fc effector functions via Fc engineering of mAbs, with a specific emphasis on the so‐called ...
  47. [47]
    Improving effector functions of antibodies for cancer treatment - PMC
    We have recently developed a successful combination composed of ADCC-enhancing modification by the fucose depletion from Fc-linked oligosaccharides and CDC- ...Missing: afucosylation | Show results with:afucosylation
  48. [48]
    Dual Fc optimization to increase the cytotoxic activity of a CD19 ...
    Aug 31, 2022 · Different Fc engineering strategies have been described to improve C1q binding of monoclonal antibodies to enhance CDC (26). For example ...<|control11|><|separator|>
  49. [49]
    Complement Deficiencies Limit CD20 Monoclonal Antibody ...
    May 2, 2014 · We therefore evaluated whether complement defects in CLL patients reduced the induction of complement-dependent cytotoxicity (CDC) by using anti-CD20 MAbs.
  50. [50]
    Enhanced CDC of B cell chronic lymphocytic leukemia cells ...
    Jun 28, 2017 · Because previous studies have suggested that there are complement deficiencies in B-CLL patients that may limit rituximab efficacy, [19] the ...
  51. [51]
    Human CD59 inhibitor sensitizes rituximab resistant lymphoma cells ...
    Complement depletion by cobra venom factor or C1q-deficiency significantly reduces the antitumor activity of rituximab in mouse models(6, 13–15).
  52. [52]
    Rituximab infusion promotes rapid complement depletion and acute ...
    Our in vitro studies indicate that at high cell densities, binding of rituximab to human CD20(+) cells leads to loss of complement activity and consumption of ...Missing: activation | Show results with:activation
  53. [53]
    Factors Influencing Infusion-Related Reactions Following Dosing of ...
    Finally, while complement activation occurs rapidly after rituximab infusion and the level of complement activation is correlated with IRR severity, it does ...<|control11|><|separator|>
  54. [54]
    Exploring complement-dependent cytotoxicity by rituximab isotypes ...
    Jun 20, 2022 · The aim of this study was to systematically evaluate factors influencing complement-mediated tumor cell killing by RTX.Missing: principles | Show results with:principles
  55. [55]
    The CDC crossmatch in the era of flow cytometric cross-match ... - NIH
    The complement-dependent cytotoxicity crossmatch (CDC-XM), a technique that uses T and B lymphocytes to detect donor-specific antibodies (DSA)
  56. [56]
    Detection of HLA Antibodies in Organ Transplant Recipients - Frontiers
    The principle of the test involves incubating donor cells with recipient serum and then adding a fluorescein-labeled second anti-human immunoglobulin antibody ...
  57. [57]
    Development of the Crossmatch Test in Kidney Transplantation Up ...
    The introduction of the crossmatch (XM) test was marked by the complement-dependent cytotoxicity (CDC) method, developed by Terasaki and colleagues in 1964 [16] ...
  58. [58]
    evaluation of flow cytometry as a replacement for ... - Transplantation
    Background: Flow cytometry (FC) is increasingly being used as a crossmatch procedure in addition to the standard complement-dependent cytotoxicity (CDC) test.
  59. [59]
    Antibody‐dependent cell cytotoxicity: immunotherapy strategies ...
    Feb 21, 2017 · Antibody-dependent cellular cytotoxicity (ADCC) is a set of mechanisms that target cells coated with IgG antibodies of the proper subclasses ...Abstract · Role of the NK cell-mediated... · Cytokines to enhance NK cell...
  60. [60]
    NK Cell-Mediated Antibody-Dependent Cellular ... - Frontiers
    These results suggest that these mAbs are acting through antibody-dependent cell-mediated cytotoxicity (ADCC) by the cells that express those FcRs. In ...
  61. [61]
    Obinutuzumab-mediated high-affinity ligation of FcγRIIIA/CD16 ... - NIH
    This modification substantially enhances the binding affinity to FcγRIIIA/CD16, thus improving antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody ...
  62. [62]
    Complement System Part II: Role in Immunity - Frontiers
    Complement activation leads to opsonization and phagocytosis by C3b deposition, bacterial lysis by C5b–9 complex formation and inflammation by recruitment ...
  63. [63]
    Purified complement C3b triggers phagocytosis and activation of ...
    Jan 6, 2023 · We show that purified C3b can potently trigger phagocytosis and killing of bacterial cells via Complement receptor 1.
  64. [64]
    Antibody drug conjugate: the “biological missile” for targeted cancer ...
    Mar 22, 2022 · Antibody–drug conjugate (ADC) is typically composed of a monoclonal antibody (mAbs) covalently attached to a cytotoxic drug via a chemical linker.
  65. [65]
    CAR T Cells: Engineering Immune Cells to Treat Cancer - NCI
    Feb 26, 2025 · CAR T-cell therapy involves genetically engineering a patient's own T cells (red) to attack cancer cells (red and blue).
  66. [66]
    Evolution of anti-CD20 monoclonal antibody therapeutics in oncology
    In complement-mediated cytotoxicity (CDC), rituximab activates complement cascade and generates membrane attack complexes and as a result induce cell death. MOR ...Programmed Cell Death... · Fc-Fc Gamma Receptor... · Table 1<|control11|><|separator|>