* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download Zhang Study
Pathophysiology of multiple sclerosis wikipedia , lookup
Immunosuppressive drug wikipedia , lookup
Autoimmune encephalitis wikipedia , lookup
Adoptive cell transfer wikipedia , lookup
Sjögren syndrome wikipedia , lookup
Multiple sclerosis signs and symptoms wikipedia , lookup
Cancer immunotherapy wikipedia , lookup
The Pharmacogenetics behind Polymorphisms of the Immunoglobin G Fragment C Receptor and Efficacy of Monoclonal Antibody Treatments for Different Cancers. “Can a patient’s DNA sequence be holding the answer to if their cancer treatment will be successful? Pharmacogenetics provides the key…” Thesis - September-October 2010 By Andriana (Andrea) Karambela, 3317331 CGDB Masters Program Supervised by Dr. M.P.J.K. Lolkema, UMC Utrecht, Oncology Department Table of contents ABSTRACT ................................................................................................................ 4 1. INTRODUCTION .................................................................................................... 5 1.1 MAb Cancer Therapy: Modes of Action ....................................................................................................... 5 1.1.1 Signal Transduction Interference as an Anti-tumor Mechanism 1.1.2 ADCC and CDC May be Responsible for Efficacy of Some mAb Treatments 5 5 1.2 Treatment of Cancers with mAbs .................................................................................................................. 9 1.2.1 mAbs with Targets on Solid Tumors 1.2.2 mAbs with Lymphoma Targets 1.2.3 mAbs with Targets in the Tumor Stroma 1.2.4 mAbs with Ligand Targets 9 10 10 10 1.3 Evaluating Treatment Outcomes ................................................................................................................. 11 2. RESULTS ............................................................................................................. 13 2.1 Population frequencies of FcγR polymorphisms ........................................................................................ 14 2.2 FcγRIIa-131 polymorphisms and mAb efficacy ......................................................................................... 14 2.3 FcγRIIIa-158 polymorphisms and mAb efficacy ........................................................................................ 15 2.4 FcγRIIb-232 polymorphisms and mAb efficacy ......................................................................................... 17 2.5 FcγRIIIa-818 and FcγRIIa-535 polymorphisms and mAb efficacy .......................................................... 17 3. DISCUSSION ....................................................................................................... 19 Frequency of FcγR Genotypes in Populations .................................................................................................. 19 FcγRIIIa-158 Tumor response doesn’t necessarily correlate with increased Clinical Benefit ..................... 19 Response and PFS V/V and V-carrier Lymphoma and Breast Cancer Patients Display Better Responses Conflicting results concerning Cetuximab-Treated mCRC Rarely Does Better Response Also Correlate with Better PFS High Threshold for ADCC-induction? 19 19 20 20 21 FcγRIIa-131 ......................................................................................................................................................... 21 Polymorphism Usually does not Correlate with Response PFS Stable Disease May Be a Better Indicator of Longer PFS than CR/PR Confounding Effects of Using Combination Treatments FcγRIIb-232 Polymorphism may not Influence mAb Efficacy 21 22 22 23 23 FcγRIIIa-818A/A Cetuximab-treated CRC Patients More Likely to Have Longer PFS Response is a Reliable Read-out? Why Does FcγRIIIa-158 Genotype Correlate with Response in Lymphoma Patients but not Leukemia? 23 24 24 4. CONCLUSION ..................................................................................................... 25 SUPPLEMENTAL DATA ......................................................................................... 26 Table of Experimental Designs .......................................................................................................................... 26 FcγRIIIa-158 ........................................................................................................................................................ 27 Weng Study (2003) Zhang Study (2010) Farag Study Bibeau Study Zhang Study (2007) Paez Study Cartron Study 27 27 27 27 28 28 28 FcγRIIa-131 ......................................................................................................................................................... 28 Weng Study (2003) Mitrovic Study Kim Study Dornan Study Zhang Study (2007) Paez Study Musolino Study 28 29 29 29 29 30 30 FcγRIIb-232 ......................................................................................................................................................... 30 Musolino Study (2008) Weng Study (2009) 30 30 FcγRIIIa-818 and FcγRIIa-535 .......................................................................................................................... 31 Pander Study 31 LITERATURE CITED ............................................................................................... 32 Abstract The vertebrate immune system has evolved to become particularly good at protecting an organism from extrinsic pathogens such as bacterial and fungal infections. Today the cutting edge of medicine is trying to manipulate the immune system of cancer patients into attacking their own cancerous cells. This can be facilitated through the use of monoclonal antibody (mAb) treatments targeted towards tumor cell receptors or ligands that are responsible for the flourishing of particular types of cancers. It has been postulated that mAb treatments may work by stimulating the patient’s immune system to kill cancerous cells through antibody-dependent cell-mediated cytotoxicity (ADCC). This process may be responsible in part for the efficacy of these antibody treatments. Curiously, the response of tumors to mAbs varies amongst patients. One hypothesis for this stratification amongst patients is that polymorphisms within immune cell receptor genes can influence the affinity of the immune cells to the mAb and hence may control how well the immune system attacks cancer cells. Our study focuses on the relationship between the efficacy of monoclonal antibodies currently used in the clinic for treatment of solid and hematological cancers and polymorphism of the immunoglobin G fragment C receptor. Using 14 studies as evidence, it appears that there is not a clear cut answer for a relationship between FcγR polymorphisms and efficacy of mAb treatment. Perhaps this is due to the many different kinds of combinational therapies which are used in the clinic. However the trend seems to be that the FcγRIIIa-158V/V genotype does improve response to treatment and can provide a longer progression-free survival (PFS) of lymphoma and breast cancer patients. For the FcγRIIa-131 polymorphism, it appears H-carriers can have a longer median PFS than other patients regardless of tumor response status. We did find that mAb treatments do not correspond with FcR polymorphisms in patients with chronic lymphocytic leukemia. Furthermore we find that “stable disease” classification can be linked with longer PFS and could therefore potentially be considered a good prognostic indicator for clinical benefit. Further research needs to be done, potentially with larger patient cohorts and proper controls to come to a definitive answer on if the FcR polymorphisms are a useful predictor of tumor response to mAb treatment. 1. Introduction 1.1 MAb Cancer Therapy: Modes of Action Monoclonal antibodies (mAb) are an effective means to treat many different types of cancer. Our study investigates if a cancer patients’ genotype for particular polymorphisms significantly changes their response to or enhances their survival after mAb treatment. 1.1.1 Signal Transduction Interference as an Anti-tumor Mechanism Some mAb treatments are effective due to their ability to block target function. Targets have been specifically chosen as they contribute to tumor growth or sustainability, an example being growth factor receptors. Use of mAbs targeting such receptors can attenuate tumor growth and can even make the tumor cells sensitive to chemotherapeutic drugs. At the molecular level these mAbs act by blocking activating ligands from binding to receptors and sterically preventing dimerization of the receptor, and in some cases can even promote cellular internalization of the receptor, like the mAb cetuximab (Sunada et al., 1986; Li et al., 2005). Alternatively mAbs can act by blocking ligand-bound receptors from forming dimers with other receptors which normally causes activation; such is the mechanism of the mAb pertuzumab (Franklin et al., 2004). While signal transduction interference may be the primary mode of action of mAbs, there may exist others. 1.1.2 ADCC and CDC May be Responsible for Efficacy of Some mAb Treatments Besides blocking target function, mAb may also promote the killing of tumor cells by activation of the immune system. Antibody Structure Antibodies are heterodimers made of two light chains and two heavy chains (Strome et al., 2007). Complementarity-determining regions are the amino terminus of each heavy and light chain, and are comprised of variable amino acid sequences that give the antibody its target specificity. The rest of the antibody contains relatively little variation in sequence and is called the constant region (Fc). Originally therapeutic mAb were of murine origin and their efficacy was not as high as expected, as human immune responses to the murine antibody produced minimal beneficial effects. However with the creation of chimeric mAbs which grafted the human Fc region onto the murine backbone containing the variable region, this problem was counter-acted and the efficacy of mAb treatments became higher (Adams and Weiner, 2005). Humanized mAb accomplished similar results but instead used a human mAb backbone and grafted on the murine variable regions. Most therapeutic mAb are humanized using the IgG1 isotype as a backbone. The Fc region of this immunoglobin can evoke a response of the human immune system to fight the cancer, namely antibody-dependent cell-mediated cytotoxicity (ADCC), as well as complement-dependent cytotoxicity (CDC). ADCC Antibody-dependent cell-mediated cytotoxicity (ADCC) is thought to be able to influence the immune system’s adaptive response against tumor cells through therapeutic mAb treatments (Adams and Weiner, 2005). The first step in this process is the binding of the mAb to a tumor cell antigen. A Natural Killer cell (NK) can then bind through its Fc receptor to the Fc region of the bound-mAb. Once receptors on the NK cell are crosslinked, this initiates a cascade that releases granzymes and perforin which lyse the tumor cell. The tumor cell debris is taken up by antigen-presenting cells. Once presented to B cells, antibodies can be produced against numerous antigens of the tumor. Also, cytotoxic T lymphocytes can be targeted to the tumor antigens and facilitate killing of the tumor cells. This process causes an immediate destruction of tumor cells with latent anti-tumor effects caused by adaptive immunity. This process can be mediated through the binding of the mAb to the receptor of the effector cell. It has been thought that polymorphisms of the FcRs can be an important regulator of tumor shrinkage if they interfere with the affinity of the receptor for the mAb, which will be expanded upon later. Complement-dependent Cytotoxicity Another immune system pathway capable of killing tumor cells is complementdependent cytotoxicity (CDC). It is also thought to be partially induced through the use of certain mAb treatments. IgG1 and IgG3 can probably activate this system through the complement activation pathway (Adams and Weiner, 2005). The pathway is initiated when the mAb binds to the tumor cell and a conformation change in the structure of the mAb allows for CDC proteins to bind to it and the CDC cascade begins. Ultimately the cascade ends with the formation of the membrane attack complex (MAC). The MAC is capable of creating holes within the membrane of the tumor cell allowing water and solutes to flow freely in and out leading to the tumor cell’s demise. One study shows that the CDC pathway does contribute to efficacy in vivo of rituximab treatment. They show that mice inoculated with lymphoma cells were not able to be cured with rituximab when the complement system was disrupted (Di Gaetano et al., 2003). Both ADCC and CDC are mediated through FcRs of effector cells. This is why polymorphisms of the FcRs has been focused upon in an effort to determine if it is a source of the variation in response amongst patient’s treated with certain mAb. Fcγ Receptors Fcγ receptors (FcγR) are important for promoting and regulating immune response to immune complexes, as the Fc region of IgG is recognized by the FcγR of effector immune cells. In humans there are three classes of FcγRs expressed by immune cells: FcγRI, FcγRII and FcγRIII. Furthermore there are “a”, “b” and “c” forms of FcγRII and “a”, “b” forms of FcγRIII. The receptors are different functionally. The activating FcγRs will incite cytotoxicity, and include FcγRIIA, FcγRIIC, FcγRIIIA and FcγRIIIB (Smith and Clatworthy, 2010). FcγRIIA and FcγRIIIA are the receptors pertinent to our investigation. FcγRIIB is the only receptor that when activated, inhibits cytotoxicity signaling and is also discussed here. A receptor’s expression is restricted to certain cell types which further discriminate the receptor’s functional specificity (Table 1). One study showed that the FcγR of effector cells is important for effectiveness of antibody treatments against tumors in mice. They tested the efficacy of trastuzumab and rituxan on eradicating human tumor xenographs in mice lacking various FcγRs. They found that the mechanism of efficacy of the mAb treatments was through minimal binding to FCgammaRIIB (anti-ADCC receptor), while preferentially binding to the FcgammaRIII (proADCC) (Clynes et al., 2000). The anti-tumor effects of the mAb treatment indeed depended upon the FcγR-mediated response in mice at least. To find out if this mechanism is relevant in the clinic is the purpose of our study. To do so we analyze 14 studies investigating the relationship between efficacies of mAb treatments in relation to polymorphisms of the FcγR. Table 1 - Expression of some FcγR (adapted from Smith and Clatworthy, 2010) Receptor Activating FcγRIIA FcγRIIIA NK cell Lymphoid Inhibitory FcγRIIB B cell Plasma cell Monocyte Expression Myeloid DC Platelet Macrophage Neutrophil Granulocyte Basophil Mast cell The FcγRIIIa-158V/F polymorphism was most commonly evaluated in studies comparing mAb therapy and FcR polymorphism (13 studies). FcγRIIIa-158 is in the membrane proximal domain receptor structure and is generally thought be located within the IgG-binding site (Koene et al., 1997). The receptors on NK cells with a valine (V) amino acid at this position are better at binding to IgG than those with a phenylalanine (F). Cells extracted from heterozygotic patients (V/F) showed intermediate binding levels, an indication of a gene-dosing effect. The clinical implications of this may mean that V-carriers have a better ability to clear pathogens as their receptors will bind more IgG than F/F patients. On the other hand, perhaps in vivo the V polymorphism enhances the binding of the receptor to cytophilic IgG and therefore decreases availability of the receptors, preventing receptor/immune complex interaction. For similar reasons FcγRIIa-131 is the next most commonly studied polymorphism (11 studies). The binding of FcγRIIa to murine IgG1 diminishes with the presence of a histidine (H) at position 131 while binding is restored in receptors with an arginine at this position (R) (Warmerdam et al., 1991). Although this study did not test human IgG1 or IgG3 which would give us a more accurate idea of what actually happens in vivo. Lastly, we analyzed two studies that observe a polymorphism of the inhibitory receptor FcγRIIb and effectiveness of mAb treatment. The genotype T/T at position 232 is found more often in patients with systemic lupus erythematosus (SLE) than healthy individuals (Kyogoku et al., 2002). SLE is an autoimmune disease where inflammation a key problem. This has led to the hypothesis that the 232I genotype is contributing to proper inhibitory signaling of the immune cells which may be impaired in T/T SLE patients. Unfortunately only a few studies looked at these polymorphisms in patients with the same type of cancer. The nature of the different kinds of cancers and modes of action of the different mAbs can help us better understand the results of these varied studies. 1.2 Treatment of Cancers with mAbs Targets of therapeutic mAbs can be on the surface of solid tumor cells or circulating malignant cells. The targets can be part of the cancer cell’s membrane (i.e. transmembrane receptors), receptor ligands (i.e. vascular growth factors), or even parts of the tumor stroma, like extracellular matrix proteins. 1.2.1 mAbs with Targets on Solid Tumors Trastuzumab (Herceptin ®; Genentech, Inc., South San Francisco, CA) is a humanized mAb targeted to the extracellular domain of HER2/neu (See Table 2). HER2/neu (c-erbB-2) is a member of the epidermal growth factor receptor (EGFR) family. It has been found to be over-expressed in roughly 25% of breast cancers, inducing a cellular proliferation and apoptosis resistance. Table 2 - Unconjugated Monoclonal Antibodies used in Cancer Therapy Generic Name (Trade Name) Trastuzumab (Herceptin) Rituximab (Rituxin) Cetuximab (Erbitux) Bevacizumab (Avastin) Alemtuzumab (Campath-1H) Panitumumab (ABX-EGF) Origin Humanized Chimeric Chimeric Chimeric Humanized Human mAb Isotype Human IgG1 Human IgG1 Human IgG1 Human IgG1 Human IgG1 Human IgG2 Target HER2/neu CD20 EGFR VEGF CD52 EGFR Indication Breat Cancer Lymphoma Colorectal cancer (CRC) CRC, Lung cancers Chronic lymphocytic leukemia Non-small cell lung, CRC Year approved by FDA 1998 1997 2004 2004 2001 2006 Cetuximab (Erbitux; ImClone Systems Inc, Branchburg, NJ) is a chimeric IgG1 mAb targeted at EGFR (Table 2). It acts by preventing the ligand from binding to the receptor, thwarting downstream proliferation signaling cascades. Studies have shown that cetuximab interferes with the signal transduction but it can also illicit ADCC or CDC in vitro and hence has multifaceted anti-tumor activity (Kang et al., 2007). Cetuximab is effective against EGFR-over-expressing cancers either on its own or in combination with chemotherapeutic drugs like irinotecan (Sobrero et al., 2008). Panitumumab is also an anti-EGFR mAb but it has a higher affinity for EGFR than cetuximab and hence is a stronger inhibitor. Comparatively it also has less side severe side effects than cetuximab as it lacks murine sequences. In treatment of colon cancer both drugs have similar efficacy (Yang et al., 1999). 1.2.2 mAbs with Lymphoma Targets Rituximab is a CD20 mAb (Rituxan®; Genentech, Inc., South San Francisco, CA). The mechanism behind this mAb mode of action is ADCC, CDC, and apoptosis as shown by in vitro studies (Shan et al., 2000). Rituximab is effective against previously treated low-grade B cell lymphomas (Bernstein et al., 1988; McLaughlin et al., 1998), low-grade or follicular non-Hodgkin lymphoma with relapsed or primary refractory disease (Piro et al., 1999). Alemtuzumab is a CD52-targeted mAb that can be used to treat chronic lymphocytic leukemia (CLL). However it can cause higher toxicities than Rituximab and can cause serious immunological complications, therefore it has been used less in the clinic (Christian and Lin, 2008). As leukemia affects circulating cells instead of a stationary solid tumor mass this may add an extra variable into our study. 1.2.3 mAbs with Targets in the Tumor Stroma The tumor stroma can be a good place to look for targets for mAb cancer treatments as tumor blood vessels, fibroblasts, inflammatory cells and matrix proteins found here differ from those in normal tissue. However these mAb have not been evaluated yet in regards to FcR polymorphism and are therefore not included in our study. 1.2.4 mAbs with Ligand Targets Targeting ligands of receptors can be an effective means to fight tumors by blocking tumor progressing signal transduction pathways from being activated. Bevacizumab is an anti-VEGF (vascular endothelial growth factor) targeted mAb used as treatment of many solid tumors (Willet et al., 2004). This type of mAbs will attach to the ligand of the receptor and therefore is not physically attached to the tumor cells itself. This excludes the possibility of ADCC or CDC from being the cause of tumor response or extended survival, therefore ligand-targeted mAbs were excluded from our study. 1.3 Evaluating Treatment Outcomes Clinical studies commonly consider three factors when evaluating effect of a cancer therapy; tumor response, progression-free survival (PFS) and overall survival (OS). For patients affected with solid tumors, the response in size of the tumor to a treatment is evaluated in everyday practice as well as in clinical trials. It can be determined by two different methods. Using a computed tomography (CT) scan, the diameter of the tumor can be established. The World Health Organization (WHO) uses bidimentional readings from CT scans (sum of the products of bidimentional readings of each tumor) to determine response, while the Response Evaluation Criteria in Solid Tumors (RECIST), calls for simplified unidimentional CT readings (sum of the longest diameter of each tumor). Both methods yield comparable results in determining tumor response (Park et al., 2003). There is a classification system based on degree of tumor response. Complete response (CR) is defined identically by both WHO and RECIST: disappearance of tumor(s) confirmed at 4 weeks. Partial response (PR) is defined by both as a reduction in tumor size of at least 65% volume-wise, confirmed at 4 weeks. Stable disease (SD) is defined as the observation of neither PR nor PD. However RECIST is less stringent then WHO on progressive disease (PD) classification: RECIST requires a tumor volume increase of at least 73% in verses WHO which requires only a 40% increase (Padhani and Ollivier, 2001). RECIST chose a higher threshold as to minimize the effect of enlarging small tumors. A common set of definitions for response concerning non-Hodgkin’s lymphomas comes from the Cheson guidelines: complete response defined as at least 75% reduction in sum of the products of the longest diameters of the tumors (SPD) (Cheson et al., 1999). Partial response is defined as at least 50% decrease in SPD. Stable Disease defines as less than a PR response but not progressive disease (PD), while PD is characterized as at least a 50% increase in SPD or the appearance of new lesions. Tumor shrinkage is a traditional factor evaluated in phase II clinical trials because it is relatively quick way to assess if a drug has anti-tumor properties. In the past a response to a drug tended to reflect increased patient survival (PFS and OS) which is usually evaluated in phase III. This conjecture has held true for cytotoxic drugs. However recently it has been pointed out that a number of effective molecularly targeted cancer drugs have shown marginal impact on response rate but subsequently showed strong positive results concerning PFS and OS (Dhani et al., 2009). These studies indicate that tumor stabilization may be an important indicator of an effective treatment resulting in clinical benefit, more so than observing a CR or PR. OS is a clinical end-point of most importance and is considered the “gold-standard” measurement for testing new cancer drugs (Dhani et al., 2009). However this measurement is subject to confounding factors such as effects of crossover designs or the use of salvage therapies. Only a couple of the FcR polymorphism studies have been able to look at this parameter. PFS is a middle of the road end-point. It avoids the confounding effects of OS like crossovers or subsequent treatments. The relevancy of PFS is determined by the intervals of follow-ups. Although PFS is under the influence of potential assessment and investigator bias if the trail is not blinded. Our investigation focused on the studies evaluating the relationship between FcγR polymorphisms and efficacy of non-conjugated, cell-targeted mAb therapies for solid tumors and hematological cancers. We take into account if the studies are retrospective or prospective, cohort size and previous patient treatments, how polymorphisms were detected, frequency of polymorphism, means of measuring response, response itself, timing of evaluations and progression-free survival (PFS). Tumor response and PFS were the primary endpoints of our study. Tumor shrinkage was as it is more a reflection of ADCC or CDC being activated than PFS or OS. We chose to look at PFS as well as it is a good indicator of clinical benefit and OS was simply not available in most cases. Knowing if the FcγR polymorphisms are a biomarker for outcome of mAb treatment would be a great help in selectively treating only the patients who would most likely benefit. This would assist in avoiding chemotherapy toxicity and reducing treatment costs. 2. Results Seeing as ADCC is responsible for part of the efficacy of some mAb treatment, researchers have hypothesized that polymorphisms of the FcR gene that alter affinity of the receptor for the mAb can influence the ADCC response to a tumor. This may alter the PFS of patient. Ultimately if the FcR polymorphisms can be used as a biomarker for response to mAb treatments, this would help in choosing to only treat patients that would most likely benefit from the mAb. This would assist in avoiding chemotherapy toxicity and reducing treatment costs. Most studies on the association of FcRs with mAb efficacy have so far focused on two polymorphisms: (1) FcγRIIa (CD 32) polymorphism at position 131, histidine (H)/ arginine (R) (van Sorge et al., 2003) and (2) FcγRIIIa (CD16) polymorphism at position 158, valine (V)/phenylalanine (F). As FcRIIa-131H and FcγRIIIa-158V have the higher binding affinities to IgG1 than the R or F variants respectively. Therefore it is hypothesized that patients who are carriers for 131H and/or 158V have the favored genotypes for initiating the anti-tumor ADCC mechanism (Parren et al., 1992; Koene et al., 1997; Strome et al., 2007). Furthermore we were expecting to see a gene-dosing effect where heterozygotes of the favored allele would have an intermediate response/PFS while the homozygotes, H/H or V/V, would have the best outcomes. 2.1 Population frequencies of FcγR polymorphisms Each study stated the number of patients with each genotype (Table 3). This allows confirmation that there is not linkage between the polymorphism and the disease. Only the Musolino study found a population frequency out of Hardy-Weinberg equilibrium. They didn’t compare this genotype frequency to that of a control group so it cannot be certain that the genotype is not linked the disease. Table 3 – Frequency of genotypes of various FcγR polymorphisms. Yellow indicates populations not in Hardy-Weinberg equilibrium Disease, Study Follicular Lymphoma, Weng 2003 B-cell Lymphoma, Zhang 2010 B-cell Lymphoma, Mitrovic 2007 B-cell Lymphoma, Kim 2006 T-NHL, Zhang 2010 CLL, Dornan 2010 CLL, Lin 2005 CLL, Farag 2004 CRC, Bibeau 2009 CRC, Zhang 2007 CRC, Paez 2010 Breast Cancer, Musolino 2010 Follicular Lymphoma, Cartron 2002 Frequency of Genotypes (Percent) FcgRIIIa-158 V/V V/F F/F 15 46 39 32 53 15 28 55 17 5 48 47 21 71 7 10 50 40 12 30 58 20 40 40 15 63 22 14 40 46 15 39 45 20 48 32 20 45 35 CRC, Pander 2010 FcRIIIa-818 A/C or A/A C/C 47 53 CRC, Pander 2010 FcRIIa-535 A/G or A/A G/G 27 73 Disease, Study Follicular Lymphoma, Weng 2003 B-cell Lymphoma, Mitrovic 2007 B-cell Lymphoma, Kim 2006 CLL, Dornan 2010 CLL, Lin 2005 CLL, Farag 2004 CRC, Bibeau 2009 CRC, Zhang 2007 CRC, Paez 2010 Breast Cancer, Musolino 2010 Follicular Lymphoma, Cartron 2002 Frequency of Genotypes (Percent) FcRIIa-131 H/H H/R R/R 23 49 28 40 46 14 56 37 7 26 55 19 18 67 15 20 57 23 28 44 28 26 48 26 26 52 22 18.5 63 18.5 29 51 20 FcRIIb-232 Breast Cancer, Musolino 2010 I/I 81 2.2 FcγRIIa-131 polymorphisms and mAb efficacy The H allele of the FcγRIIa-131 locus confers a higher binding affinity of the receptor for IgG. Therefore it was expected that H-carriers or H/H patients would have better responses and longer PFS than the other patients. We found that there was only one study out I/T T/T 11 8 of 10 studies that found a significant relationship between genotype and response (Table 4). The H/H patients showed the better response in that study which agrees with our hypothesis. Furthermore there were two studies which found a non-significant trend for H/H or H-carrier patients with better responses. However there were also two studies which found that Rcarriers have a non-significant trend towards better response. Interesting these studies were both looking at colorectal cancer patients treated with cetuximab plus chemotherapy. Perhaps these insignificant trends simply show up as the cohorts are too small (See Supplemental Data Table). Overall this evidence makes a very weak case for the FcγRIIa-131 polymorphism being linked to response in general, but there is a significant relationship for follicular lymphoma treated with Rituximab. Eight studies examined the correlation between the FcγRIIa-131 polymorphism and length of PFS. Three of them found that patients with the H/H genotype or H-carriers have significantly longer average PFS than the other patients (Table 4). These three studies are each looking at different diseases, each treated with a different drug. This shows that while significant connections between genotype and response as well as PFS is rare (only one study out of 10 found this), it is more common to find a good clinical benefit associated with the genotype and simultaneously not find tumor response. Follicular lymphoma treated with rituximab appears to have significant association with response and PFS. 2.3 FcγRIIIa-158 polymorphisms and mAb efficacy Concerning the FcγRIIIa-158 polymorphism the V-carriers were expected to have better response than other patients, as this allele makes the receptor have a higher affinity for the IgG. We found that the V/V genotype conferred a better response over the F-carriers in 4 of the 12 studies, and one study showed V-carriers had the advantage (Table 4). No study found F/F or F-carriers to hold an advantage for response. Interestingly 4 of the 5 studies which found the V-carriers to have better responses were performed in lymphoma patients (Bcell and follicular) treated with rituximab. In general it appears that there is a significant link between the V-genotype and response for lymphoma and breast cancer patients. Looking at non-significant trends, three of the 12 studies show that F-carriers have better responses than the other patients. This is unexpected and counter-intuitive as it has been shown that the F-allele has a lower affinity for the mAb. Two of these studies were on patients with colorectal cancer (CRC). Table 4 – Response and PFS According to Genotype of FcRs. FcRIIa-131 Response H/H H/R PFS Disease, Study Treatment Follicular Lymphoma, Weng 2003 CRC, Zhang 2007 Rituximab Cetuximab + I H/H advantage No Association # R/R H/H advantage H-carriers advantage H/H H/R R/R Breast Cancer, Musolino 2010 B-cell Lymphoma, Mitrovic 2007 Trastuzumab + T Rituximab + CHOP No Association + No Association H/H advantage over R/R No Association B-cell Lymphoma, Kim 2006 CRC, Bibeau 2009 Rituximab + CHOP Cetuximab No Association * No Association No Association No Association CRC, Paez 2010 CLL, Dornan 2010 Cetuximab/Pan ~I Rituximab + FC No Association # No Association No Association No Association CLL, Lin 2005 CLL, Farag 2004 Alemtuzumab Rituximab No Association No Association N/A N/A FcgRIIIa-158 Response V/V V/F PFS Disease, Study Treatment Follicular Lymphoma, Cartron 2002 Follicular Lymphoma, Weng 2003 Rituximab Rituximab V/V advantage V/V advantage F/F No Association V/V advantage V/V V/F F/F Breast Cancer, Musolino 2010 B-cell Lymphoma, Kim 2006 B-cell Lymphoma & T-NHL, Zhang 2010 CRC, Bibeau 2009 CRC, Zhang 2007 B-cell Lymphoma, Mitrovic 2007 CLL, Dornan 2010 Trastuzumab + T Rituximab + CHOP Rituximab + CHOP Cetuximab + I Cetuximab Rituximab + CHOP Rituximab + FC V/V advantage V/V advantage V-carriers advantage No Association No Association † No Association No Association V/V advantage No Association No Association ‡ V/V advantage F-carriers advantage No Association No Association CRC, Paez 2010 CLL, Lin 2005 CLL, Farag 2004 Cetuximab/Pan ~I Alemtuzumab Rituximab No Association † No Association No Association † No Association N/A N/A FcRIIb-232 Response Disease, Study Treatment Breast Cancer, Musolino 2010 Follicular Lymphoma, Weng 2009 Thrastuzumab + T Rituximab I/I I/T PFS T/T No Association º No Association I/I I/T T/T No Association N/A FcRIIIa-818 Response Disease, Study Treatment CRC, Pander 2010 Cetuximab + COB A/A PFS A/C or C/C N/A A/A A/C or C/C A/A advantage FcRIIa-535 Response Disease, Study Treatment CRC, Pander 2010 Cetuximab + COB A/A N/A A/G or G/G PFS A/A A/G or G/G No Association Legend: + I = irinotecan, ~ I = some patients received irinotecan, Pan = 12% of patients received Panitumumab instead, T = Taxane, FC = Fludarabine + Cyclophosphamide, † F-Carriers trend advantage (Non-Sig), ‡ V- Carriers trend advantage (Non-Sig), * H-Carriers trend advantage (Non-Sig), # R-Carriers trend advantage (NonSig), + H/H advantage (Non-Sig), N/A - data not available, COB = Capecitabine, Oxaliplatin and Bevacizumab, º I/I adv (not sig.) In conclusion the V-allele is a good indicator of response for lymphoma and breast cancer patients, while potentially the F-allele is linked to better response in CRC patients. Of 5 the studies that found the V/V or V-carrier patients to be significantly better responders, only 2 of them found that these patients also had a longer PFS. Interestingly these 2 studies were evaluating responses from different mAb, Rituximab and Trastuzumab, illustrating that this is not a drug-specific effect. Furthermore these two studies were looking at patients with different diseases, follicular lymphoma and breast cancer, implying this effect is also not specific to cancer type. There were 2 studies which did not find a correlation of genotype with response but did find a correlation with PFS. One found that the V/V carriers had the advantage and the other found that the F-carriers did. Strangely both studies were on CRC patients. In conclusion there is some evidence that the FcγRIIIa-158V/V genotype corresponds with better PFS for patients with follicular lymphoma or breast cancer. There is inconclusive evidence for linkage of genotype with the PFS of patients with CRC. 2.4 FcγRIIb-232 polymorphisms and mAb efficacy Two studies evaluated a polymorphism of the inhibitory receptor FcγRIIb. The T allele may weaken the affinity of the receptor for IgG. As it is an inhibitory receptor we thought the T allele would enhance ADCC/CDC and result in a better response/PFS. Both Follicular lymphoma patients and breast cancer patients showed responses to rituximab and trastuzumab respectively which did not correlate their FcγRIIb-232 genotype (Table 4). In the Musolino study (2008) there is evidence that the T-carriers are less likely to respond to the trastuzumab treatment than I/I patients, but it was not significant (see Supplemental Data). Furthermore PFS was not significantly associated with any genotype in both studies (Weng study could not evaluate PFS for T/T patients). It appears that this receptor and/or polymorphism do not play a significant role in the efficacy of mAb treatments. 2.5 FcγRIIIa-818 and FcγRIIa-535 polymorphisms and mAb efficacy A recent randomized study performed by Pander and colleagues examined the FcγRIIa-535 and FcγRIIIa-818 polymorphisms in KRAS wildtype mCRC patients (Pander et al., 2010). Previously untreated patients were administered chemotherapy (capecitabine + oxaliplatin) and bevacizumab with the addition of cetuximab. Response was not evaluated in this study. The FcγRIIIa-818 C-carriers were found to be associated with decreased PFS (Table 4). Most important was the finding that the FcγRIIIa-818 polymorphisms were not associated with PFS in patients not given cetuximab. This highlights that the polymorphism is not causing a non-drug related effect on clinical outcome. FcγRIIa-A535G polymorphism was found to not be associated with PFS. In conclusion the FcγRIIIa-818 A/A genotype appears to be linked with increase PFS for CRC patients. 3. Discussion We set out to prove or disprove if the higher affinity of certain FcγR polymorphisms for IgG correlate with better response and longer PFS of cancer patients treated with therapeutic mAb. Frequency of FcγR Genotypes in Populations In general the frequencies of genotypes in the populations appear to have normal distributions (Table 3). An exception is the Kim study which used Korean patients and seems to have a population biased towards certain genotypes which could skew their correlation results. For this reason the Kim study results which show a significant correlation of the FcγRIIIa-15V/V genotype with response but not with PFS may be the result of this particular population. Although the Zhang study used patients of a different population with same disease, treated with the same mAb and found similar results. On the other hand there is the Mitrovic study which used Caucasian patients and found no correlation with either response or PFS, a plausible explanation for the difference in results with the Kim study. Weather the difference between populations really is the cause of the differing results is unknown, but perhaps the low number of patients in certain genotypes in the Kim study is causing results to be insignificant which may become significant in a larger population or vice versa. FcγRIIIa-158 Tumor response doesn’t necessarily correlate with increased Clinical Benefit Response and PFS Concerning the FcγRIIIa-158 polymorphism, 5 studies of 12 studies found that the V/V genotype (the higher affinity allele) or V-carriers had better response over the F-carriers. Of these 5 the studies, only 2 found that these patients also had a longer PFS. Perhaps the disparity between response and PFS in three of the studies occurs as the drug affects tumor size, but the side-effects it causes are the trumping factor and limit PFS. V/V and V-carrier Lymphoma and Breast Cancer Patients Display Better Responses Importantly, 4 of the 5 studies which found the V-carriers to have better responses were performed in lymphoma patients (B-cell and follicular) treated with rituximab. In general it appears that there is a significant link between the V-genotype and response for lymphoma and breast cancer patients. Conflicting results concerning Cetuximab-Treated mCRC There were 2 studies which did not find FcγRIIIa-158 genotype correlating with response but did find a correlation with PFS. One of them found that the V/V patients had longer PFS while the other found that the F-carriers did. Both of these studies were treating CRC patients with cetuximab. A major difference between the two was that the Bibeau study used cetuximab plus irinotecan as treatment (V/V patients advantage) and the Zhang study (2007) used single-agent cetuximab as treatment (F-carriers advantage). Perhaps this variable causes the difference, but it could also be that the Zhang study simply had too small a cohort (39 patients) get a clear result, while the Bibeau study was larger (69 patients) and perhaps is thus more reliable. Furthermore there was a third study (Paez, 2010) which also looked at CRC patients treated with Cetuximab, but no correlation with response or PFS was found. This study by far had the largest patient cohort, 104 patients, and therefore is probably the more definitive study out there. In conclusion it appears that there is not agreement on the link between FcγRIIIa-158 genotype and response/PFS concerning CRC. Rarely Does Better Response Also Correlate with Better PFS Of 5 the studies that found the V/V or V-carrier patients to be significantly better responders, only 2 of them found that these patients also had a longer PFS. Interestingly these 2 studies were evaluating responses from different mAb, Rituximab and Trastuzumab, illustrating that this is not a drug-specific effect. Furthermore these two studies were looking at patients with different diseases, follicular lymphoma and breast cancer, implying this effect is not specific to cancer type. In conclusion there is some evidence that the FcγRIIIa-158V/V genotype corresponds with better PFS for patients with follicular lymphoma or breast cancer. Our study found that there is not convincing evidence for better response being linked to longer PFS in general (10 of the 12 studies). A plausible explanation may be that the patients that had significantly longer PFS was because they had stable disease (rather than PR or CR), and this may be an overlooked mechanism of improving clinical benefit. For example, some studies have pointed out that a non-response does not always signify a shorter PFS or OS. In a retrospective study by Grothey and colleagues (2008) it was found that the classifications “responder” or “non- responder” put upon mCRC patients after bevacizumab + IFL treatment did not correspond with those who had longer PFS and OS. This could be why we find little correlation with response and those patients who had longer PFS. Indeed this is the case in the Zhang study (2007). Concerning their FcγRIIIa-158 polymorphism results, 71% of F/F patients showed PR or SD and 29% PD, while heterozygote F/V patients showed 71% PR or SD and 29% PD, while the V/V homozygous patients showed 20% PR or SD and 80% PD. Seeing as F-carriers had significantly longer PFS than V/V patients it can be said that PR + stable disease is an indicator of clinical benefit. The stable disease classification could be seen as a positive indicator of clinical benefit, a predictive condition that is currently not given enough credit. High Threshold for ADCC-induction? Another question lingering in the wake of these results is why the studies which found the V/V genotype at an advantage concerning response, did not find a significant intermediate response for V/F patients. This would be expected if the hypothesis that the V-allele is advantageous because of its heightened affinity, then wouldn’t we expect to see a gene-dose effect, instead of the clear advantage of the V homozygotes. Perhaps there is a threshold of activated receptors that needs to be reached to have a positive effect and this threshold is simply not reached in heterozygotes. In general it appears that there is a significant link between the V-genotype and response for lymphoma and breast cancer patients. Furthermore there is some evidence that the FcγRIIIa-158V/V genotype corresponds with better PFS for patients with follicular lymphoma (FL) or breast cancer. There is inconclusive evidence for linkage of genotype with the PFS of patients with CRC. Perhaps this genotype is useful only for evaluating treatment options of patients with FL or breast cancer. FcγRIIa-131 Polymorphism Usually does not Correlate with Response Concerning the FcγRIIa-131 polymorphism, there was one study out of 10 studies that found a relationship between genotype and response. H/H patients showed better response in that study which agrees well with the fact that the H-allele confers higher affinity of the receptor for the mAb. However it is not so surprising that less studies found correlation with this polymorphism those of the FcγRIIIa-158 polymorphism, as the affinity of the FcγRIIa receptor is strong for murine IgG1 and human IgG2, which can be quite different from the human IgG1 backbone which most mAb tested here are. PFS 3 of the 8 studies looked at the polymorphism and PFS found correlation, H/H or Hcarriers having the advantage. These three studies are each looking at different diseases, each treated with a different drug, which is quite remarkable, implying again that this phenomenon is not drug- or disease-specific. Stable Disease May Be a Better Indicator of Longer PFS than CR/PR A number of effective molecularly targeted cancer drugs have shown marginal impact on response rate then subsequently showed strong positive results concerning PFS and OS (Dhani et al., 2009). These studies indicate that tumor stabilization may be an important indicator of an effective treatment resulting in clinical benefit, as we proposed with the FcγRIIIa-158 polymorphism. There is evidence for the FcγRIIa-131 polymorphism acting similarly. The Zhang study (2007) shows that the 131R polymorphism is not significantly associated with response. However of the H/H patients 0% had a PR, 78% SD and 22% PD, while H/R patients showed 6% PR, 71% SD and 24% PD, and R/R patients showed 14% PR, 0% SD, and 86% PD. If one only takes into account percentage of PR, it shows the R-carriers have an (not significant) advantage response-wise, which is what Zhang notes, but if one looks at the PR + SD it is the H-carriers who have a great advantage, nearly five times as high percentage over the R/R patients. This correlates with the significantly longer PFS of the H-carriers. It appears that the R/R genotype certainly points to a poor prognosis as they have shorter median PFS and higher PD percentages. As Zhang used Cetuximab mono-therapeutically, I think the data from this study a more solid result than those from studies using combinational therapies which may be confounding results. The Zhang study makes a strong case that cetuximab can stabilize a tumor can extend PFS. Confounding Effects of Using Combination Treatments The Dornan study (2010) showed no influence of genotype over response or PFS concerning both FcRIIa-131 and FcγRIIIa-158 polymorphisms. This could be due to the combination treatment administered to patients, FC + rituximab. They do point out that the difference in PFS is significant between heterozygous patients for both FcRIIa-131 and FcγRIIIa-158 treated with R-FC compared to the heterozygous patients treated only with FC. This implies this second part of the therapy (FC) may have confounding effects on the interpretation of the results. This could also be a factor of many of the other studies seeing as combinational therapies are common practice, but can add in an extra variable into the equation. Furthermore the Dornan study also had a relatively low occurrence of V/V patients in their study and perhaps evaluating more V/V patient would change the significance. FcγRIIb-232 Polymorphism may not Influence mAb Efficacy The two studies examining the FcγRIIB-232 polymorphism found no association of genotype with response of PFS. This may be because it is an inhibitory receptor, and by having a polymorphism that make the receptor bind less well to the mAb apparently does not provide an advantage, such as indirectly promoting inflammation by its prevention of activating the inhibitory signaling pathway. FcγRIIIa-818A/A Cetuximab-treated CRC Patients More Likely to Have Longer PFS The polymorphisms FcγRIIa-535 and FcγRIIIa-818 were looked at exclusively by one study on CRC patients. Only FcγRIIIa-818A/A patients were shown to have a significantly longer PFS than the other patients. Seeing as the studies concerning the FcγRIIIa-158 polymorphism and CRC had highly conflicting results, the Pander experiment should also be repeated with another cohort, as perhaps CRC is a very variable disease concerning response, and obviously the results from one experiment are not decisive. Interestingly the Pander study made sure that their patient cohort was genotyped to be KRAS-wildtype. Perhaps this is a variable future CRC studies should also take into account. Response is a Reliable Read-out? Using response in tumor size as an end-point may be a difficult reliable read-out, as measurements of tumor diameters can fall to error and observer variation in determining lesion boundaries is high (Padhani and Ollivier, 2001). This can lead to mis-classification of disease stage and confound results, and could be why there was a lack of significant effects found in many of the studies. PFS would also include the necessary volume determination procedure and would be subject to such error. Due to this perhaps OS is a better indicator of clinical benefit as it is a measurement not given to such inaccuracy. Overall, there needs to be more studies performed which evaluate larger numbers of patients, are prospective and take into account OS. Why Does FcγRIIIa-158 Genotype Correlate with Response in Lymphoma Patients but not Leukemia? CLL cells have lower expression of CD20, the target for rituximab, than follicular or B-cell lymphomas (Dornan et al., 2010). This could explain why 4 of the 5 lymphoma studies we reviewed state a relationship with the FcR polymorphisms and tumor response to rituximab while none of the 3 studies on CLL found an association. If the rituximab target is expressed less in CLL then hypothetically ADCC would probably occur less and hence an imperceptible difference in treatment efficacy amongst the FcR genotypes of CLL patients would result. Furthermore, it has been proposed that an over-expression of CD55 or CD59 can cause inhibition of CDC (Bannerji et al., 2003). Perhaps this is the case for CLL and explains why the 3 studies on this disease and rituximab show no correlation with the polymorphisms. Alternatively it may be possible that ADCC is better at eliminating neoplastic B-cells in lymph nodes (Lymphoma) than when they’re in the blood and bone marrow (CLL) (Farag et al., 2004). This can also cause the discrepancy between results over these two disorders. Rituximab and alemtuzumab probably work through non-ADCC/CDC mechanisms in patients with CLL. It has been postulated that efficacy of the CDC pathway in tumor degeneration may be dependent upon number or density of antigen sites on a given tumor cell’s surface (Adams and Weiner, 2005). This idea has been further supported by an in vitro study which shows CDC activation is correlated with expression levels of CD20 (Golay et al., 2001). Furthermore the CDC pathway can help to contribute to ADCC because upon release of C3a and C5a, NK and other effector cells can be recruited to the tumor and promote ADCC (Gelderman et al., 2004). We hypothesize that because CLL cells have lower levels of CD20 they are not affected by ADCC/CDC and therefore no correlation with the FcR polymorphism are found. For CLL treatment, target-disrupting effects are therefore probably the main mode of action. 4. Conclusion In conclusion it appears that there is not a clear cut answer for relationship between FcγR polymorphisms and efficacy of mAb treatment. Perhaps due to the many different kinds of combinational therapy that are used, a clear cut answer many never be found. Each study was slightly different concerning patient cohort or exact treatment. However the trend seems to be that FcγRIIIa-158V/V genotype does improve response to treatment and can provide a longer PFS for lymphoma and breast cancer patients. For the FcγRIIa-131 polymorphism, it appears H-carriers can have longer PFS than other patients. mAb treatments do not correspond with FcR polymorphisms in patients with follicular lymphoma. Furthermore we find that the “stable disease” classification can be linked with longer PFS and could potentially be considered a good prognostic indicator for clinical benefit. Supplemental Data Table of Experimental Designs Disease, Study Follicular Lymphoma, Cartron 2002 Previous Treatment Treatment Dose 72 patients chemotherapy (10 bone marrow transplants) Rituximab 4 Weekly 375 mg/m2 Rituximab 4 Weekly 375 mg/m2 - Thrastuzumab + T 4 mg/kg, then 2 mg/kg - Rituximab + CHOP 6 Cycles 375 mg/m2 Failed 1 irinotecan treatment, majority also oxalipatin Failed 2 chemotherapy regimens Rituximab + CHOP 21-day cycle (6-8 cycles) Cetuximab + I Standard + I Cetuximab Standard Rituximab + CHOP 3 Cycles (plus 1-5 more) 375 mg/m2 Rituximab + FC N/A CRC, Paez 2010 Previous Treatment unnamed Failed 1 chelotherapy regimen Cetuximab/Pan ~I N/A CLL, Lin 2005 ~3 treatments Alemtuzumab week 1: 3-30 mg, then thrice every week (12 weeks) CLL, Farag 2004 - Rituximab 4 Weekly 375 mg/m2 CRC, Pander 2010 Follicular Lymphoma, Weng 2009 - Cetuximab + COB 400 mg/m2 , then 250 mg/m2 81 patients relapsed Rituximab 4 Weekly 375 mg/m2 Follicular Lymphoma, Weng 2003 Breast Cancer, Musolino 2010 B-cell Lymphoma, Kim 2006 B-cell Lymphoma & TNHL, Zhang 2010 CRC, Bibeau 2009 CRC, Zhang 2007 B-cell Lymphoma, Mitrovic 2007 CLL, Dornan 2010 Disease, Study Follicular Lymphoma, Cartron 2002 Follicular Lymphoma, Weng 2003 Breast Cancer, Musolino 2010 B-cell Lymphoma, Kim 2006 B-cell Lymphoma & T-NHL, Zhang 2010 Number DNA of Tissue Patients Origin Genotyping Method Evalutation Criteria Evaluation TimePoints after end of Treat 49 BM/PB RD Cheson day 50 or 78, 1 year 87 T/PB RD+DS Cheson month 1/3, then every 3 months 54 PB rtPCR +/or DS RECIST N/A RD Cheson 2/3 cycle, month 1, then every 4 months 113 BM/PB 48 PB AS-PCR Cheson every 2 cycles CRC, Bibeau 2009 69 T/N AS-PCR or DS RECIST every 2-3 months CRC, Zhang 2007 39 PB RD or AS-PCR Own Criteria every 6 weeks B-cell Lymphoma, Mitrovic 2007 58 N/A PCR+RD Cheson cycle 3/4, then at end of treatment CLL, Dornan 2010 210 PB RT-PCR+RD N/A N/A CRC, Paez 2010 104 PB 48.48 Dynamic Array RECIST every 2-3 months CLL, Lin 2005 36 PB PCR+RD Cheson/NCI 96 Weekly CLL, Farag 2004 30 PB PCR+RD Cheson month 2 CRC, Pander 2010 127 PB AS-Probes + RT-PCR RECIST every 3 cycles Follicular Lymphoma, Weng 2009 101 T/PB AS-Probes Cheson month 1/3, then every 3 months Legend : BM/PB PCR+RD T DS AS-PCR Bone Marrow or Peripheral Blood PCR + Allele-Specific Restrection Enzyme Digest Tumor Direct Sequencing Multiplex allele-specific PCR rt N reverse transcription of cDNA no previous treatment normal tissue FcγRIIIa-158 Weng Study (2003) The FcγRIIIa-158 V/V genotype was determined to be a predictive factor for response at 1 to 3 months, 6, 9 and 12 months. About 45% of patients with the V/V genotype had PFS at 2 years post-treatment, while 12% of heterozygotic patients had PFS, and 16% for the F/F patients. Zhang Study (2010) A retrospective study by Zhang and colleagues (2010) analyzed patients with B-cell lymphoma (34 patients) and T cell non-hodgkin lymphoma (14 patients). All patients received rituximab plus CHOP (21-day cycle for 6-8 cycles). Patients who reached neither PR or relapsed received a salvage treatment of etoposide + cisplatin + cytarabine + methylprednisolone (ESHAP)/mitoxantrone + ifosfamide + etoposide (MINE) for 2-4 courses until PR was reached. The rituximab + CHOP was effective (PR + CR) in 82% of patients with the V/V genotype. 83% of heterozygous (V/F) patients showed a response to treatment, while patients homozygous F had only 60% had a good response rate. While the researchers state they found no statistically significantly differences amongst the genotypes concerning PFS or OS, the PFS at 1 year of V/V patients was 73%, heterozygotes (V/F) 69% and F/F homozygotes 20%. The F/F patients appear to be at the disadvantage. Only response rate was deemed by the researchers as an independent prognostic factor of OS, not genotype. Farag Study In a study by Farag and colleagues (2004) the efficacy of rituximab on CLL patients was evaluated considering the FcγRIIIa-158 polymorphism. 33% of V/V patients responded positively, 42% of V/F and 50% of F/F patients. The trend implies a benefit for the F-carriers but no significant association with response was found. Bibeau Study Patients homozygous 158V had 6.9 months PFS compared to the F-carriers who had 3.2 months.When taking into account the FcγRIIa and FcγRIIIa polymorphisms together, no significant difference in response was observed between 131H and/or 158V homozygotes versus the others. However 131H and/or 158V homozygotic patients did have significantly longer PFS compared to the others (5.5 months vs. 3 months). The results of the Bibeau study showed that combined the FcγRIIa-131H/H and/or FcγRIIIa-158V/V polymorphisms are prognostic factors for PFS in mCRC patients treated with cetuximab plus irinotecan. However they are not influential concerning tumor responses. Zhang Study (2007) Concerning the FcγRIIIa-158V/F polymorphism results from the Zhang study (2007), 14% of the F/F patients showed PR, 57% SD and 29% PD, while heterozygote F/V patients showed 0% PR, 71% SD, and 29% PD, while the V/V homozygous patients showed 0% PR, 20% SD and 80% PD. This trend shows that the F-carriers have a much better response to cetuximab than the patients homozygous for 158V, but it is not a significant association. Concerning PFS the Zhang study found that patients who were homozygous for FcγRIIIa-158F had a median PFS of 2.3 months while heterozygous patients had a PFS of 2.4 and the homozygous 158V patients had a PFS of 1.1 months. Combining both FcRIIa and FcγRIIIa genotypes yielded highly statistically significant relationship between the polymorphisms and PFS. Patients who are carriers of both H and F allele(s) have a median PFS of 3.7 months while R/R or V/V patients have a PFS of 1.1 months on average. Paez Study While it is not significant, there is a trend of benefit in response for the F-carriers (54% of V/V patients showed PR or SD, 60% of V/F and 72% of F/F patients). Cartron Study PFS survival was evaluated 3 years after treatment. 56% of V/V patients had PFS and 35% of F-carriers, not a significant difference. FcγRIIa-131 Weng Study (2003) An advantage of the H/H patients was also found when observing PFS: 37% of H/H patients had PFS as 2 years post treatments, while only 13% of H/R patients and 19% of R/R patients. The H/H PFS was significantly longer than that of the other genotypes. These results were confirmed again in a more recent study by the same investigators expanding the same patient pool from 87 to 101 patients (Weng and Levy, 2009). Again the H/H patients had better response and better PFS than the R-carriers. Mitrovic Study In a study by Mitrovic and colleagues (2007) diffuse large b-cell lymphoma (DLBCL) patients were evaluated for their response to treatment of rituximab plus standard CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisolone). Patients who responded to the treatment were continued on it for another 1-5 cycles, while patients who did not respond were either given radiation treatment and/or second-line chemotherapy followed by peripheral blood stem cell transplantation. Kim Study In a retrospective study of Korean DLBCL patients Kim and colleagues (2006) investigated response to first-line R-CHOP therapy in relation to their FcγR polymorphisms. 76% of these patients received only the R-CHOP treatment while the remaining 33% of patients were subsequently treated with radiation. A median of 6 cycles of R-CHOP had been administered to patients, rituximab given at the standard dose. Event free survival (EFS) is defined as time from day 1 of the first R-CHOP cycle until treatment failure, which includes DP, recurrence or death from any cause. The Kim study (2006) found there was a non-significant trend of better response towards patients with the H/H genotype (ORR of 95% compared to 92% of H/R and 75% of the R/R patients). Dornan Study This retrospective study revealed a slight trend towards the H/H genotype of being more beneficial for median PFS (31.8 months for H/H, 27.3 for H/R and 26.7 for R/R patients). Zhang Study (2007) These patients were part of a phase II open-label multicenter study. Treatment was continued until disease progression or the occurrence of toxicity. An objective response was defined as reduction of at least 50% of tumor size using CT. Objective responses were confirmed 4 weeks after the initial detection. Tumor progression was defined as an increase of at least 25% of tumor area or the appearance of new lesions. Response was also measured retrospectively in an independent blinded assessment. The Zhang study shows a trend for R-carriers having an advantage can be found considering PR alone. Concerning PFS, the patients that were 131H homozygous had a PFS of 2.4 months, while heterozygotic patients had a median PFS of 3.7 months, and the 131R homozygous patients had a PFS of 1.1 months. This would indicate that the H-carriers have the advantage concerning PFS. Paez Study When considering the FcγRIIa-131 polymorphism they found 54% of H/H patients had PR or SD, 64% of H/R patients and 77% of R/R. This implies a trend of R-carriers to have better response to this treatment. Perhaps with a larger cohort this would be significant. Musolino Study This study comprised of patients with HER2/neu-positive metastatic breast cancer that were treated with trastuzumab plus taxane (paclitaxel or docetaxel) as a first-line therapy and 34 controls who were patients with metastatic breast cancer but not selected for HER2/neu amplification, and were only administered taxane without trastuzumab. Patients homozygous for the FcγRIIa-131H allele showed 70% positive response, while heterozygous patients (H/R) were 44% positive, and 40% of 131R homozygous patients responded positively. This difference is not significant but there is a trend which shows that R-carriers are less likely to respond to the trastuzumab treatment. FcγRIIb-232 Musolino Study (2008) 55% of patients with the FcγRIIb-232I/I genotype were positive responders, of the heterozygotic patients (I/T) 17% were responders and of the homozygous 232T patients 25% were responders to trastuzumab. Found to be not significant. Weng Study (2009) Patients with the FcγRIIb-232I/I genotype had similar responses (64%) to the treatment compared to patients with the I/T (67%) genotype or the T/T (50%) at 1 to 3 months posttreatment. There was also no significant difference at 6, 9 nor 12 months. About 20% of patients with the I/I genotype had PFS at two years compared to the 43% of I/T patients, this was not significant. FcγRIIIa-818 and FcγRIIa-535 Pander Study A randomized study with KRAS wildtype mCRC patients cohort (Pander et al., 2010). Patients were part of a phase III CAIRO2 study. A cycle of treatment consisted of cetuximab being administered at 400 mg/m2 on the first day and then 250 mg/m2 each week thereafter for 3 weeks. The FcγRIIIa-818 C-carriers had a median 8.2 months PFS compared to patients with the 818A/A genotype of 12.8 months. Literature Cited Adams GT and Weiner LM (2005). Monoclonal antibody therapy of cancer. Nat Biotechnol, 23, 1147-57. Bannerji R, Kitada S, Flinn IW, Pearson M, Young D, Reed JC and Byrd JC (2003). Apoptotic-regulatory and complement-protecting protein expression in chronic lymphocytic leukemia: relationship to in vivo rituximab resistance. J Clin Oncol. 21,1466-1471. Baselga, J. (2001). Clinical trials of HerceptinR (trastuzumab). Eur J Cancer 37, S18–S24. Bernstein, N. et al. Association of serum Rituximab (IDEC-C2B8) concentration and antitumor response in the treatment of recurrent low-grade or follicular non-Hodgkin’s lymphoma. Ann. Oncol. 9, 995–1001 (1988). Bibeau F, Lopez-Crapez E, Di Fiore F, Thezenas S, Ychou M, Blanchard F, Lamy A, PenaultLlorca F, Frebourg T, Michel P, et al. (2009). Impact of Fc{gamma}RIIa-Fc{gamma}RIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus rinotecan. J Clin Oncol. 27, 1122-9. Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, and Watier H (2002). Theraputic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood 99, 754-8. Cheson BD, Horning SJ, Coiffier B, Shipp MA, Fisher RI, Connors JM, Lister TA, Vose J, Grillo-Lopez A, Hagenbeek A et al. (1999). Report of an international workshop to standardize response criteria for non-Hodgkin’s lymphomas. J Clin Oncol 17, 1244. Christian BA and Lin TS (2008). Antibody therapy for CLL. Semin Hemotol. 45, 95-103. Cobleigh, M, Vogel CL, Tripathy D, Robert NJ, School S, Fehrenbacher L, Wolter JM, Paton V, Shak S, Lieberman G, Slamon DJ (1999). Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J.Clin. Oncol. 17, 2639–2648. Coiffier, B. Rituximab in the treatment of diffuse large B-cell lymphomas. Semin. Oncol. 29, 30–35 (2002). Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, Bets D, Mueser M, Harstrick A, Verslype C et al. (2004). Cetuximab monotherapy and Cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 351, 337-45. Dhani N, Tu D, Sargent DJ, Seymour L, and Moore MJ (2009). Alternate endpoints for screening phase II studies. Clin Cancer Res. 15, 1873-82. Di Gaetano N, Cittera E, Nota R, Vecchi A, Grieco V, Scanziani E, Botto M, Introna M, Golay J (2003). Complement activation determines the therapeutic activity of rituximab in vivo. J. Immunol. 171, 1581–1587. Dornan D, Spleiss O, Yeh RF, Duchateau-Nguyen G, Dufour A, Zhi J, Robak T, Moiseev SI, Dmoszynska A, Solal-Celigny P et al. (2010). Effect of FCGR2A and FCGR3A variants on CLL outcome. Blood, Aug 12. Fan, Z., Masui, H., Altas, I. & Mendelsohn, J. Blockade of epidermal growth factor receptor function by bivalent and monovalent fragments of 225 anti-epidermal growth factor receptor monoclonal antibodies. Cancer Res. 53, 4322–4328 (1993). Farag SS, Flinn IW, Modali R, Lehman TA, Young D and Byrd JC (2004). Fc gamma RIIIa and Fc gamma RIIa polymorphisms do not predict response to rituximab in B-cell chronic lymphocytic leukemia. Blood 103, 1472-4. Franklin, MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM, Sliwkowski MX (2004). Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell 5, 317–328. Gelderman, K.A., Tomlinson, S., Ross, G.D. & Gorter, A. Complement function in mAbmediated cancer immunotherapy. Trends Immunol. 25, 158–164 (2004). Golay J, Lazzari M, Facchinetti V, Bernasconi S, Borleri G, Barbui T, Rambaldi A, Introna M (2001) CD20 levels determine the in vitro susceptibility to rituximab and complement of Bcell chronic lymphocytic leukemia: further regulation by CD55 and CD59. Blood 98, 3383– 3389. Grothey A, Hedrick EE, Mass RD, Sarkar S, Suzuki S, Ramanathan RK, Hurwitz HI, Goldberg RM and Sargent DJ (2008). Response-Indepdendent Survival Benefit in Metastatic Colorectal Cancer: A Comparative Analysis of N9741 and AVF2107. J Clin Oncol 26, 183-9. Haga, Y., Sivinski, C., Woo, D. & Tempero, M. (1994). Dose related comparison of antibodydependent cellular cytotoxicity with chimeric and native monoclonal antibody 17–1A. Improved cytolysis of pancreatic cancer cells with chimeric 17–1A. Int. J. Pancreatol. 15, 43–50. Hartung G, Hofheinz RD, Dencausse Y, Sturm J, Kopp-Schneider A, Dietrich G, FacklerSchalbe I (2005). Adjuvant therapy with edrecolomab versus observation in stage II colon cancer: a multicenter randomized phase III study. Onkologie 28, 347–350. Kang et al. (2007). High affinity Fc receptor binding and potent induction of antibodydependent cell-mediated cytotoxicity in vitro by anti-epidermal growth factor antibody cetuximab. J Clin Oncol 25, 3712-3718. Kim DH, Jung HD, Kim JG, Lee JJ, Yang DH, Park YH, Do YR, Shin HJ, Kim MK, Hyun MS and Sohn SK (2006). FCGR3A gene polymorphisms may correlate with response to frontline R-CHOP therapy for diffuse large B-cell lymphoma. Blood 108, 2720-5. Koene HR, Kleijer M, Algra J, Roos D, von dem Borne AE, and de Haas M (1997). Fc gammaRIIIa-158V/F polymorphism influences the binding of the IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype. 90, 1109-14. Kyogoku C, Dijstelbloem HM, Tsuchiya N, Hatta Y, Kato H, Yamaguchi A, Fukazawa T, Jansen MD, Hashimoto H, van de Winkle et al. (2002). Fcgamma receptor gene polymorphisms in Japanese patients with systemic lupus erythematosus: contribution of FCGR2B to genetic susceptibility. Arthritis Rheum 46, 1242-54. Li S, Schmitz KR, Jeffery PD, Wiltzius JJ, Kussie P (2005). Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell 7, 301–311. Lin TS, Flinn IW, Modali R, Lehma TA, Webb J, Waymer S, Moran ME, Lucas MS, Farag SS and Byrd JC (2005). FCGR3A and FCGR2A polymorphisms may not correlate with response to alemthuzumab in chronic lymphocytic leukemia. Blood 105, 289-91. McLaughlin, P. et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J. Clin. Oncol. 16, 2825–2833 (1998). Miller, R, Maloney, D, Warnke, R & Levy, R (1982). Treatment of B-cell lymphoma with monocolonal anti-idiotype antibody. N. Engl. J. Med. 306, 517–522. Mitrovic Z, Aurer I, Radman I, Ajdukovic R, Sertic J and Labar B (2007). FCgammaRIIIA and FCgammaRIIA polymorphisms are not associated with response to rituximab and CHOP in patients with diffuse large B-cell lymphoma. Haematologica 92, 998-9. Musolino A, Naldi N, Bortesi B, Pzzuolo D, Capelletti M, Missale G, Laccabue D, Zerbini A, Camisa R, Bisagni G, et al. (2008). Immunoglobin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol. 26, 1789-96. Norton L, Slamon D & Leyland-Jones B (1999). in Proceedings of the American Society of Clinical Oncology vol. 18, 127a, ASCO Publications, Alexandria, Virginia, USA. Padhani AR and Ollivier L (2001). The RECIST (Response Evaluation Criteria in Solid Tumors) criteria: implications for diagnostic radiologists. Br J Radiol. 92, 179-81. Paez D, Pare L, Espinoza I, Salazar J, Del Rio E, Barnadas A, Marcuello E and Baiget M (2010). Immunoglobin G fragment C receptor polymorphisms and KRAS mutations : Are they useful biomarkers of clinical outcome in advanced colorectal cancer treated with antiEGFR-based therapy? Cancer Sci, May 18. Pander J, Gelderblom H, Antonini NF, Tol J, van Krieken JH, van der Straaten T, Punt CJ (2010). Correlation of FCGR3A and EGFR germline polymorphisms with the efficacy of cetuximab in KRAS wild-type metastatic colorectal cancer. Eur J Cancer 46, 1829-34. Park JO, Lee SI, Song SY, Kim K, Kim WS, Jung CW, Park YS, Im YH, Kang WK, Lee KS and Park K (2003). Measuring response in solid tumors: comparison of RECIST and WHO response criteria. Jpn J Clin Oncol 33, 533-7. Parren PW, Warmerdam PA, Boeije LC, Arts J, Westerdaal NA, Vlug A, Capel PJ, Aarden LA, van de Winkel JG (1992). On the interaction of the IgG subclasses with the low affinity Fc gamma RIIa (CD32) on human monocytes, neutrophils, and platelets. J Clin Invest. 90, 1537-46. Piro LD, White CA, Grillo-Lopez AJ, Janakiraman N, Saven A, Beck TM (1999). Extended Rituximab (anti-CD20 monoclonal antibody) therapy for relapsed or refractory low-grade or follicular non-Hodgkin’s lymphoma. Ann. Oncol. 10, 655–661. Saltz LB, Meropol NJ, Loehrer PJ, Needle MN, Kopit J, and Mayer RJ (2004). Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J Clin Oncol. 22, 1201-8. Shan, D., Ledbetter, J. & Press, O. (2000) Signaling events involved in anti-CD20-induced apoptosis of malignant human B cells. Cancer Immunol. Immunother. 48, 673–683. Smith KG and Clathworthy MR (2010). FcgammaRIIB in autoimmunity and infection: evolutionary and therapeutic implications. Nat Rev Immunol 10, 328-43. Sobrero AF, Maurel J, Fehrenbacher L, Scheithauer W, Abubakr YA, Lutz MP, VegaVillegas ME, Eng C, Steinhauer EU, Prausova J et al. (2008). EPIC: phase III trial of cetuximab plus irinotecan after fluoropyrimidine and oxaliplatin failure in patients with metastatic colorectal cancer. J Clin Oncol. 26, 2311-9. Strome SE, Sausville EA and Mann D (2007). A mechanism perspective of monoclonal antibodies in cancer therapy beyond target-related effect. Oncologist. 12, 1084-95. Sunada, H., Magun, B.E., Mendelsohn, J. & MacLeod, C.L.(1986). Monoclonal antibody against epidermal growth factor receptor is internalized without stimulating receptor phosphorylation. Proc. Natl. Acad. Sci. USA 83, 3825–3829. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS (2000). New guidelines to evaluate the response to treatment in solid tumors: European Orgainization for research and Treatment of Cancer, National Cancer Institute of the US, National Cancer Institute of Canada. J Natl Cancer Inst. 92, 205-216. van Mierlo, G.J. et al. CD40 stimulation leads to effective therapy of CD40(+) tumors through induction of strong systemic cytotoxic T lymphocyte immunity. Proc. Natl. Acad. Sci. USA 99, 5561–5566 (2002). Van Sorge NM, van der Pol WL and van de Winkle JGJ (2003). FcγR polymorphisms: Implications for function, disease susceptibility and immunotherapy. Tissue Antigens. 61, 189-202. Vuist, W., Levy, R. & Maloney, D. Lymphoma regression induced by monoclonal antiidiotypic antibodies correlates with their ability to induce Ig signal transduction and is not prevented by tumor expression of high levels of bcl-2 protein. Blood 83, 899–906 (1994). Warmerdam PA, van de Winkle JGJ, Vlug A, Westerdaal NA and Capel PJ (1991). A single amino acid in the second Ig-like domain of the human Fc gamma receptor II is critical for human IgG2 binding. J Immunol. 147, 1338-43. Weng WK and Levy R (2003). Two immunoglobin G fragment C receptor polymorphisms independently predict response to rituximab in patients with folliclar lymphoma. J Clin Oncol 21, 3940-7. Weng WK, Levy R (2009). Genetic polymorphism of the inhibitory IgG Fc receptor FcgammaRIIb is not associated with clinical outcome in patients with follicular lymphoma treated with rituximab. Leuk Lymphoma 50, 723-7. Willett CG, Boucher Y, di Tomaso E, Duda DG (2004). Direct evidence that the VEGFspecific antibody bevacizumab has antivascular effects in human rectal cancer. Nat. Med. 10, 145–147. Yang, X. et al. (1999). Eradication of established tumors by a fully human monoclonal antibody to the epidermal growth factor receptor without concomitant chemotherapy. Cancer Res. 59, 1236–1243. Zhang W, Gordon M, Schultheis AM, Yang DY, Nagashima F, Azuma M, Chang HM, Borucka E, Lurje G, Sherrod AE, et al. (2007). FCGR2A and FCGR3A polymorphisms associated with cinical outcome of epidermal growth factor receptor expressing metastatic colorectal cancer patients treated with single-agent cetuximab. J Clin Oncol. 25, 3712-8. Zhang W, Wang X, Li L, Duan MH, Zhou DB (2010). Fcgamma receptor IIIA polymorphisms and efficacy of rituximab therapy on Chinese diffuse large B-cell lymphoma. Chin Med J 123, 198-202.