Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jan 2.
Published in final edited form as: Subcell Biochem. 2014;75:221–254. doi: 10.1007/978-94-007-7359-2_12

Chapter 13: Carbonic Anhydrase IX as an Imaging and Therapeutic Target for Tumors and Metastases

Narges K Tafreshi 1, Mark C Lloyd 2, Marilyn M Bui 2,3, Robert J Gillies 1, David L Morse 1
PMCID: PMC4282494  NIHMSID: NIHMS644624  PMID: 24146382

Abstract

Carbonic anhydrase IX (CAIX) which is a zinc containing metalloprotein, efficiently catalyzes the reversible hydration of carbon dioxide. It is constitutively up-regulated in several cancer types and has an important role in tumor progression, acidification and metastasis. High expression of CAIX generally correlates with poor prognosis and is related to a decrease in the disease-free interval following successful therapy. Therefore, it is considered as a prognostic indicator in oncology.

In this review, we describe CAIX regulation and its role in tumor hypoxia, acidification and metastasis. In addition, the molecular imaging of CAIX and its potential for use in cancer detection, diagnosis, staging, and for use in following therapy response is discussed. Both antibodies and small molecular weight compounds have been used for targeted imaging of CAIX expression. The use of CAIX expression as an attractive and promising candidate marker for systemic anticancer therapy is also discussed.

1. Introduction

Carbonic anhydrases (CAs) are metalloproteins, usually containing Zinc (EC 4.2.1.1) that catalyze the reversible hydration of carbon dioxide. These are widespread in nature, being found in bacteria, plants and mammals (1, 2). Sixteen CA isoforms have been characterized to date in mammals, which differ in their cellular localization and catalytic activity, susceptibility to different inhibitors, and tissue-specific distribution. Some isoforms are strictly cytosolic: (CAI, CAII, CAIII, CAVII and CAXIII), some are membrane-associated (CAIV, CAIX, CAXII, CAXIV and CAXV), some are mitochondrial (CAVA and CAVB), and one (CAVI) is secreted into saliva and milk. There are also three non-catalytic enzymes (CAVIII, CAX and CAXI), which are named as carbonic anhydrase related proteins, CARPs. Although structurally homologous, the inactivity of these CAs is due to the absence of one or more Zn-binding histidine residues (1, 36). The structure and function of CA isozymes have been extensively reviewed by Hassan et al., 2012 (7).

CAs catalyze the reversible dehydration of bicarbonate (CO2 + H2O ↔ HCO3 + H+) (1, 2, 8); Since CO2 is the main byproduct of all oxidative processes, the CAs play a fundamental role in maintaining acid-base equilibrium (1, 2, 9), and are involved in crucial physiological processes such as respiration, electrolyte balance, biosynthetic reactions that require bicarbonate as a substrate (gluconeogenesis, lipogenesis, and ureagenesis), bone resorption, and calcification (1, 2). Moreover, CAs are involved in pathological processes such as cancer progression, obesity and epilepsy and thus have been proposed as therapeutic targets.

A cDNA coding for the hCAIX protein was first cloned and investigated by Pastorek et al. (10), and the CAIX gene was further characterized by the same group (11). Isoform CAIX, originally named the “MN protein”, has been identified using a monoclonal antibody (M75) as a plasma membrane antigen detected in a human cervical carcinoma cell line, HeLa (12). The expression of MN antigen also was detected in various tumor cell lines and surgical tumor specimens, but not in the corresponding normal tissues, suggesting its potential usefulness as a tumor marker (13). Because it was the ninth mammalian CA identified, the MN protein was renamed CAIX. A renal cell cancer-associated antigen (G250) detected by mAb was then reported to be identical with MN/CAIX by another research group (14, 15).

CAIX is among the most active CAs for the CO2 hydration reaction, and contains four domains on the basis of sequence similarity (11, 16): an N-terminal proteoglycan-like (PG) domain, a CA catalytic domain, a transmembrane segment (TM), and an intracytoplasmic (IC) portion. Other CAs, such as the cytosolic CAII and the membrane-associated CAXII lack the PG-like domain (Figure 1). It is worth mentioning that the M75 mAb binds to the repetitive epitope in the PG region, allowing selective detection of CAIX without cross-reactivity with other CAs (17). The PG-like domain immediately adjacent to the catalytic domain allows the enzyme to act efficiently at acidic pH values (18, 19). It has also been proposed that the PG domain is involved to cell adhesion and tumor invasion (17, 20, 21) via its interaction with β-catenin, which leads to a reduction in E-cadherin mediated cell-cell adhesion, thus promoting cell motility and invasion (20). In contrast to the PG domain, the CAIX catalytic domain has significant sequence identity (from 30% to 40%) to catalytic domains of other hCA isozymes. Both the CA and PG domains are glycosylated (16). The cytoplasmic tail of CAIX contains three potential phosphorylation sites (T443, S448 and Y449) that can participate in signal transduction. When tyrosine 449 of CAIX is phosphorylated, it can interact with the regulatory subunit of PI3K, which can transactivate the Ser/Thr protein kinase Akt, and have a direct impact on cellular glucose metabolism (22).

Figure 1.

Figure 1

Proposed model showing the structural arrangement of the CAXII and CAIX dimer on the cellular membrane (23, 24). PG= proteoglycan-like domain, CA= catalytic domain, TM= transmembrane segment, IC= intracellular, cytosolic tail.

Acetazolamide is a clinically used sulfonamide-based CA inhibitor. An X-ray crystal structure of the catalytic domain of CAIX in complex with acetazolamide has been determined (Figure 1). The crystal structure has identified significant differences between CAIX and the other α-CA isoforms that also have solved three-dimensional structures. These developments have led to increased interest in the rational drug design of CAIX isozyme-specific CA inhibitors with potential for use as anticancer drugs for systemic therapy targeting disseminated disease (23). The CAIX protein assembles as a dimer which is stabilized by the formation of an intermolecular disulfide bond between the same Cys residue located on two CA catalytic domains (16, 23). The active site clefts and the PG domains are thus located on one face of the dimer, while the C-termini are situated on the opposite face, facilitating protein anchoring to the cell membrane (23, 24). It has been shown that neither lipid raft localization nor phosphorylation is likely required for CAIX dimerization or activity in MDA-mb-231 breast cancer cell (25).

CAIX expression is restricted to a few tissues including the epithelium of the stomach (gastric mucosa), biliary tree, crypt cells of the duodenum, and the epithelium of the small intestine (26, 27), but is overexpressed in response to tumor hypoxia in many tumor types and plays a critical role in hypoxia associated tumor acidosis and development of the metastatic phenotype. The comparison of CAIX cDNA sequences from cancer cell lines compared to normal cells does not reveal mutations or alternative splicing that would indicate a different function in cancer (27). CAIX expression is associated with a poor prognosis [reviewed by (28)] and high CAIX expression is found in lymph node and distal metastases for a broad range of cancer types [see the “CAIX expression in cancer” section below]. Hence, CAIX is being pursued as a marker for development of targeted systemic therapies and diagnostic imaging probes.

2. CAIX regulation

Expression of CAIX protein is primarily regulated at the level of transcription. The promoter of the CA9 gene contains an HRE (hypoxia responsive element) that is located immediately upstream of the transcription start site at position -3/-10. HIF-1α binds to the HRE and induces transcription of the CA9 gene in response to hypoxia and increased cell density (29). The transcriptional regulation of CAIX has been reviewed in detail recently (30). Alternative splicing of the CA9 transcript has been observed (31). Besides the predominant, full-length CA9 mRNA (1.5 kb mRNA), an alternatively spliced variant, lacking exons 8/9 among 11 exons, has been detected by RT-PCR. This mRNA variant is constitutively expressed at very low levels and codes for a truncated, cytoplasmic/secreted form of CAIX without enzymatic activity. This is very important for studies related to hypoxia-related expression of CAIX, where the presence of the alternatively spliced variant is hypoxia-independent and can provide a false-positive signal (31). Hence, primers for qRT-PCR should be carefully designed to detect the longer transcript containing exons 8/9. The physiological role of this variant is unknown. Finally, the CAIX protein can be post-translationally modified by metalloprotease mediated shedding of the ectodomain (32).

Under normoxic conditions (Figure 2, right), HIF-1α is constitutively expressed but is rapidly degraded by the ubiquitin-28S proteasome pathway (33). Specifically, two proline residues of HIF-1α (P402 and P564) and asparagine (N803) are hydroxylated by oxygen-dependent proline hydroxylases (PHD) and factor inhibiting HIF-1 (FIH-1), respectively. PHD and FIH-1 belong to the Fe(II) and 2-oxoglutarate dioxygenase superfamily. Hydroxylated HIF-1α proteins bind to the E3 ubiquitin ligase, VHL, complex, leading to its rapid degradation by the proteasome (34). Hydroxylation of N803 by FIH further inactivates HIF-1α by inhibiting its interaction with transcriptional co-activator, CBP/p300 (34).

Figure 2.

Figure 2

Hypoxia-induced gene expression mediated by the HIF1 transcription factor heterodimer. HIF-1α is degraded in normoxia by the ubiquitin-proteasomal pathway. In hypoxia, it is phosphorylated by MAPK and bound by CBP/p300, inducing heterodimirezation with HIF-1β and transcription of variety of genes.

Under hypoxic conditions (Figure 2, left), lack of O2 prevents the hydroxylation of HIF-1α by PHD and FIH (and thus degradation). Therefore, there is no VHL binding and HIF-1α is stabilized. Non-hydroxylated N803 of HIF-1α allows binding of CBP/p300 to HIF-1α and eventual translocation to the nucleus for heterodimerization with HIF-1β, producing the master transcription factor, HIF-1. Binding of HIF-1β to HIF-1α is enhanced by MAPK phosphorylation of HIF-1α (35, 36). HIF-1 then binds to target genes that contain HRE sites, including, genes broadly related to iron metabolism (e.g. erythropoietin), glucose metabolism (e.g. GLUT-1 and GLUT-3), angiogenesis (e.g. vascular endothelial growth factor), and, last but not least, genes that are involved in pH regulation (e.g. CAIX and the sodium-hydrogen exchanger, NHE-1) and induces their transcription. Additional hypoxia-induced genes are involved in cell proliferation and viability, regulation of vascular remodeling and plasticity, cell adhesion, cell matrix metabolism, and other cellular processes (29, 3537). For a detailed list of target genes, see Table 2 of Ke and Costa (38).

HIF-1α stabilization is a common observation in advanced stage invasive cancer and metastases (39) potentially by a number of mechanisms, resulting in the constitutive expression of cancer related genes, including CA9. One cancer related mechanism involves HIF-1α stabilization by glycolytic end-products, lactate and pyruvate (40), thus establishing the basis for a positive feedback loop for glycolysis and the Warburg Effect under hypoxic conditions. The tumor suppressor p53 is known to signal HIF-1α degradation as a response to DNA damage, down regulating CAIX expression (41). In most clear cell renal cell carcinomas (RCCs), the von Hippel-Lindau (pVHL) tumor suppressor is non-functional by mutation, hypermethylation or deletion. Non-functional VHL is unable to polyubiquitate HIF-1α and thus, HIF-1α is no longer recognized by the proteasomal degradation system. This leads to constitutive activation of HIF-1 even in normoxic conditions and, as a consequence, strong and uniform upregulation of CAIX and other HIF-1-induced genes (29, 4244).

Recent studies have suggested that cancer stem cells (CSCs) preferentially survive in the hypoxic niche (4547). CSCs have the ability to generate tumors and undergo the epithelial–mesenchymal transition (EMT) and are implicated in metastasis. Interestingly, a recent study showed that inhibition of CAIX activity with small-molecule inhibitors resulted in the inhibition of breast CSC expansion, leading to diminished tumor growth and metastasis (48). This result suggests that CAIX is an important therapeutic target for selectively depleting breast CSCs. The same study has identified the mTORC1 axis as a critical pathway downstream of CAIX in the regulation of CSC function and suggested that CAIX expression regulates CSC EMT by regulation of ‘stemness’ genes, such as Snail and Notch (48).

Our unpublished data and other studies have shown that expression levels of several hypoxia related proteins, including CAIX, are elevated at the invasive front of solid tumors at similar levels observed in central, perinecrotic areas (49). This invasive front is also the observed location of CSC migration (Figure 3) (50).

Figure 3.

Figure 3

Immunohistochemical staining of CAIX is an important tool to investigate 2-D localization of expression. A) Dark brown CAIX overexpression at the edge of the tumor (white arrow) contrasts not only with the adjacent microenvironment in blue (black arrow), but also with the decreased expression of the tumor >200microns from the edge (yellow arrow). B) A mask depicts regions selected by computer learning which segment and classify the tumor (red) and non-tumor (blue) regions. C) Single cell segmentation and classification of the tumor cells demonstrates the location of strong (red), moderate (orange), weak (yellow) and negative (white) expressing cells. These classifications allow investigators to quantify metrics of the IHC stain localization. Scale = 150 microns.

2.1 Cross talk between the CAs

Knockout (K/O) of CA9 in transgenic animals resulted in homozygous animals that developed gastric hyperplasia, but no changes were observed in gastric pH, acid secretion or serum gastrin levels (51). Also no tumorogenesis was observed in CA9 K/O mice, although aged CA9 null mice may suffer degenerative brain disease (52). The knockout of CA9 in mice interestingly led to the up-regulation of CAII, while expression of CAIX is upregulated about 2 fold in CA2 null mice (53). Chiche and his colleagues also observed that knockdown of CA9 in LS174Tr colon carcinoma cells led to partial compensation by up-regulation of CAXII in vitro and in vivo (54), suggesting cross talk between CAs and that such cross talk is needed for cell pH homeostasis (51, 52). The mechanism of this specific cross talk between the CAs remains unclear (55).

3. CAIX expression in tumor hypoxia

Hypoxia can be defined as the lack of oxygen supply in a tissue such that cellular mechanisms are compromised or an adaptive response occurs. Solid tumors usually have heterogenous regions of hypoxia due to insufficient supply of oxygen by irregular and functionally defective tumor vasculature. This situation creates selective pressure in favor of tumor cells that can adapt to this microenvironmental stress, with oncogenic alterations in metabolism as a result. These adaptive changes also result in expansion of cells with more aggressive phenotypes and increased metastatic potential (56).

The importance of hypoxia has been demonstrated clinically, where it is an independent predictor of poor survival in several types of cancer. In fact, hypoxia is correlated with malignant progression and resistance to conventional chemo- and radio-therapies in many cancer types (57). This resistance is partially due to poor perfusion, which limits drug delivery and tumor penetration. Also, since chemotherapeutic agents target rapidly dividing cells, often they fail to target cells in hypoxic areas which are more slowly growing or non-proliferating. Additionally, upregulation of HIF-1 can lead to chemoresistance in tumors and metastases (e.g. HIF-1 is able to activate the multidrug resistance 1 (MDR1) gene in response to hypoxia) (58, 59). Finally, radiotherapy-based treatments which rely on the generation of oxygen super-radicals also often fail to provide effective treatment because of inadequate oxygen within the tumor mass (60).

These properties of tumor hypoxia make relevant the ability to detect hypoxic regions in order to determine which patients could benefit from adjunctive anti-hypoxia therapy and to monitor hypoxia targeted therapy response (61). Consequently, significant effort has been spent to qualify reliable hypoxia biomarkers for both diagnostic and therapeutic purposes. An exciting advance in this field has been the recent development of hypoxia-activated pro-drugs (62), which specifically target the hypoxic niche with high dose systemic chemotherapy.

Although different methods such as direct tumor pO2 measurement by microelectrode (63), PET probes based on bioreductive 2-nitroimidazoles following intravenous administration (18F-misonidazole, 18F-HX4, 18F-FAZA and 18F-EF5 [reviewed by (64)]) have been used to detect hypoxia, each of these methods have some disadvantages. For example microelectrode use is constrained by the difficulty of accessing tumors, cost, invasiveness, inter-observer variability (65), failure to distinguish necrosis from hypoxia (66) and difficulty in calibration (61). Bioreductive-based probes can be limited by poor tumor-to-background ratios and non-specific signal generated by oxygen dependent metabolism of these probes (67).

As mentioned above, at the molecular level, hypoxia inducible factor 1 (HIF-1) is up-regulated in response to oxygen deprivation, binds to the hypoxia responsive element (HRE), and controls the transcriptional activity of numerous downstream genes, such as CAIX, which has been emerged as one of the most promising endogenous markers of cellular hypoxia.

Notably, however, staining of CAIX is not in complete agreement with pimonidazole (a hypoxia marker) staining in patient tumors and human xenograft tumors in mice (6872). This difference may arise from short-lived fluctuations in tumor perfusion and slow rates of CAIX degradation (73), or that the pimonidazole threshold for labeling is <1.3% oxygen, whereas up-regulation of molecular markers such as CAIX occur at pO2 values over a larger range, 0.2–2% oxygen (74).

Also, although CAIX expression correlates with the pattern of HIF-1α expression in most regions (54), exceptions have been observed where expression of CAIX is detected in the absence of HIF-1α staining (75), and these mismatches may be informative. These observations could be due to the different half-lives of HIF-1α and CAIX expression where CAIX protein is more stable (76, 77), i.e. HIF-1α is rapidly degraded in normoxic conditions and just as rapidly stabilized in hypoxic conditions. Therefore, cells that had been hypoxic and then were reoxygenated may stain for CAIX but not HIF-1α (30). In addition, when cells or tissue have only recently become hypoxic, they may stain positive for HIF-1α but not CAIX, because those cells may have been analyzed before the full onset of CAIX expression (30). Also, the duration and severity of hypoxia identified as acute, cycling (acute hypoxia followed by rapid reoxygenation), and chronic hypoxia can have differential effects on transcription and translation of HIF clients (7880). The use of CAIX as a biomarker for imaging of hypoxia is discussed in the following section.

4. The role of CAIX in metabolism and metastasis: Acid mediated invasion

Tumor acidosis, low tumor pH, is a commonly observed consequence of dysangiogenesis and increased metabolism, which are ‘hallmarks’ of cancer. Acidosis results from enhanced rates of glucose fermentation (conversion to lactic acid) and the inefficient removal of acidic end products because of deficient vasculature in the tumor mass (8183). Tumor acidosis has been reported to favor metastasis (84). Therefore it is considered an attractive therapeutic target.

Since intracellular (pHi) regulation controls multiple cellular functions such as energy production, proliferation and migration, tumor cells must develop strategies to regulate their pHi to survive and proliferate (8587). In fact, tumor cells have highly sophisticated mechanisms involving a variety of proteins and buffering systems that lead to maintenance of a moderately alkaline pHi in tumor cells while generating a markedly acidic extracellular environment, allowing them not only to survive and proliferate in hypoxia, but also favoring metastasis [reviewed by (88, 89)].

As described above, HIF-1 is activated under hypoxic conditions and in many cases it is constitutively activated in tumor cells. HIF-1 acts to regulate pH homeostasis by up-regulating expression of membrane located transporters, exchangers, pumps and ecto-enzymes such as Na+/H+ exchanger (NHE-1), H+-ATPases and monocarboxylate transporters (MCTs), that actively export acids from tumor cells, Cl/HCO3 exchangers (AE) that make the cytoplasm alkaline by pumping the weak base HCO3 into the cells, and carbonic anhydrases (CAs) (83, 9093).

CAs alone can contribute to pH regulation, i.e. raising intracellular pH and lowering extracellular pH. Intracellular CAs can mediate the conversion of intracellular HCO3- (a product of AE) into CO2. This process consumes an intracellular proton. The resulting CO2 then diffuses through the plasma membrane possibly through an aquaporin channel, wherein it is reconverted back into bicarbonate and protons by the extracellular carbonic anhydrases, e.g. CAIX, particularly in the context of a hypoxic microenvironment (94). Thus, the net effect is the movement of a proton from the cytosol to the extracellular space. The produced protons contribute to extra-cellular acidosis, leading to aggressive tumor phenotype by promoting invasion and metastasis (95). The newly generated HCO3 ions can then be transported back into the tumor cells by HCO3 transport proteins. The coupled transport process is probably essential for hypoxic cancer cells to buffer and maintain intracellular pH at near neutral conditions which is necessary for their biosynthetic reactions (26, 30, 54, 93, 94, 96). It has been shown that knockdown of CAIX minimizes the gradient in extracellular pH that occurs across three dimensional tumor spheroids, which normally have an acidic core, with a corresponding decrease in the intracellular pH of hypoxic tumor cells (97).

As mentioned previously, the crystal structure of the CAIX catalytic domain has confirmed the dimeric nature of the enzyme and showed that the N-terminal regions of both monomers are located on the same face of the dimer, while both C-termini are located on the opposite face (23). This structural organization allows for concomitant orientation of both PG domains toward the extracellular environment at the entrance to the active site clefts. This positioning of the PG domain at the border of the active site suggests a role in assisting the CA domain-mediated catalysis. In fact, the influence of the PG domain on the pH-dependent profile of the kcat/KM values for CO2 hydration was demonstrated in a recent study (23); while the CAIX catalytic domain alone presented an optimal pKa at 7.01, this value changed to 6.49 in the presence of the PG domain. Therefore, the PG domain of CAIX enables the hydration of CO2 to occur at the low pH levels observed in the tumor microenvironment (19, 98). In this context, the PG domain may act as an intrinsic buffer due to the large number of COOH side groups in this region (19).

Since an acidic microenvironment is commonly observed in cancers, methods are needed to overcome low pHe drug resistance in order to improve therapeutic efficacy (99). Targeted treatment with proton pump inhibitors (PPIs) disrupt transmembrane pH gradients and can inhibit growth in human B cell leukemia (100). Also, inhibition of CAIX catalytic activity using monoclonal antibodies or specific small molecule inhibitors leads to intracellular acidification with the consequent impairment of the tumor growth and reduced invasive capacity of the tumors (101, 102).

Alternatively, acidosis can be inhibited directly with the ingestion of alkaline buffers, such as bicarbonate (103). Alkalization of tumors by administration of bicarbonate in drinking water effectively increased the extracellular tumor pH without affecting the pHi or systemic pH and led to reduced metastases in human breast and prostate cancer mouse models and increased survival (104). Interestingly, immunohistochemical staining for CAIX in regions of ductal hyperplasia of the C57BL/6 TRAMP (transgenic adenocarcinoma mouse prostate) model showed increased expression in controls versus the group that had 200 mM sodium bicarbonate in their drinking water (105). However, these results do not generalize across all tumor cell lines, as fast-growing tumor cell lines, e.g. B-16 melanoma, showed resistance. A concern about buffer therapy is whether or not a balance can be reached between the acid load produced by the tumor and the quantity of bicarbonate that can be administrated. Thus the effectiveness of this therapy will be reduced in large tumors (106). However, the strong advantage of this strategy would be to restore a more neutral tumor pHe that should slow down the metastatic process and diminish drug resistance (77). Another approach that takes advantage of low tumor pHe involves the development by many groups of low pH activated micelle systems that penetrate into solid tumors and release their cytotoxic therapeutic payloads within the acidic microenvironment. However, additional in vivo studies are needed to determine their efficacy (107, 108).

5. CAIX expression in cancer

The use of CAIX as a prognostic indicator in oncology makes it a potentially important biomarker for the evaluation of tissues. CAIX is constitutively up-regulated in several cancer types such as the lung (109111), colon (112, 113), colorectal cancer (114), gastric cancer (115), pancreatic cancer (116), breast (117121), cervix (122125), bladder (68, 126), ovaries (127), brain (128, 129), head and neck (130132), astrocytomas (133) and oral cavity (134136). The mechanism of upregulation has been determined in some cancers, e.g. clear cell carcinoma of the kidney, where the VHL gene is mutated leading to high levels of CAIX expression (up to 150-fold) as a consequence of constitutive HIF activation (37, 137140).

High expression of CAIX has been shown to generally correlate with poor prognosis and is related to shorter disease-free and disease specific survival [reviewed (28)]. For example, CAIX is particularly highly expressed in the aggressive basal-like or triple negative breast cancers, a tumor subgroup with poor prognosis and known resistance to systemic therapy (119, 141), and high expression has been reported in patients with squamous cell carcinoma of the lung (111). However, there is still much to be uncovered regarding the prognostic indications of CAIX in various cancers. For example, it has been reported that CAIX over-expression, as detected by IHC staining, may be an indicator of a good prognosis in clear cell renal cell carcinoma (142). While, in other renal tumors such as chromophobe renal cell carcinoma and oncocytoma, CAIX is reported to be absent via IHC (143). In other cases, such as squamous cell carcinoma, IHC of CAIX does not appear to add prognostic or predictive value (144). Therefore, it is important to carefully characterize the prognostic value of CAIX in individual tumor types. Once established, the utility of CAIX expression in IHC has been shown to be a useful marker (145).

A correlation between CAIX expression and metastatic disease has been also observed. For example, a recent study that examined over 3600 human breast cancers reported that CAIX expression in distant metastases is an independent biomarker of poor prognosis (141). These results were similar to other studies (122124, 146, 147). A high level of CAIX expression in cervical cancer tumors is associated with the presence of lymph node metastases (124, 125). In addition, high stromal CAIX expression was associated with nodal metastasis and decreased survival in patients with surgically-treated oral cavity squamous cell carcinoma (148).

CAIX is also reported to be a useful IHC biomarker for the quantification of response to therapy. For example, CAIX has been used to predict renal cell carcinoma response to IL-2 therapy (149) and epirubicin/tamoxifen therapy in breast cancer (150).

Another membrane associated, CA, CAXII, is co-expressed with CAIX in several tumor tissues and highly expressed in a number of cancer types, including breast cancer (26, 151). Our group has recently reported co-expression of CAIX and CAXII in different types of breast cancer (152)(Figure 4). CAIX and CAXII staining was distributed in the cell membranes of tumor tissues. Interestingly, some samples were positive only for CAIX, some only for CAXII, and others had expression of both markers. This can be problematic when using agents that bind non-specifically to the CA active site if the goal is to target only CAIX (see discussions below). Lymph node metastases samples were positive for CAIX (71%) and CAXII (76%), both markers were expressed in 44% of the samples, and all of the positive lymph nodes were found to express either CAIX or CAXII, i.e., the combination of CAIX and CAXII covers 100% of patients with ALN metastases represented in our TMA. Heterogeneity of expression was found in 29% and 35% of the tumor samples for CAIX and CAXII, respectively, i.e. different areas of the same tumor on a histological section having different expression patterns for a given marker (152). We have planned future studies to determine if there is a correlation of CAIX expression with negative patient outcomes.

Figure 4.

Figure 4

Bubble plot of pathologist scores for CAIX and CAXII IHC staining A) for samples from all breast cancer patients on the tissue microarray (TMA), n = 180; or B) for 47 normal breast tissues, 42 ductal carcinoma in situ, 43 invasive ductal carcinomas without metastasis, 46 invasive ductal carcinomas with metastasis and 49 lymph node macrometastases of breast cancer on the TMA.

5.1 Detection of CAIX expression in tumors and metastases

The extracellular domain of CAIX, including PG and CA domains, can be released into the culture medium of CCRCC (clear cell renal cell carcinoma) cells or in the body fluids (blood, urine) of patients with CCRCC. This release is most likely a result of proteolytic cleavage. The soluble 50/54 kDa form is termed s-CAIX and it is cleared from the blood within a few days after nephrectomy (153). The concentration of s-CAIX is very low in healthy subjects (153, 154). Therefore, it can be assayed by ELISA in serum, plasma and tissue for clinical detection and prognostic evaluation of patients with different types of cancers. ELISA could be a potential quantitative method for non-invasive diagnosis of renal tumors. Two different groups have reported similar values for serum s-CAIX in CCRCC (114.0–126.1 pg/mL), higher than in other types of renal cancer and healthy samples (143, 154). Serum CAIX levels were significantly higher in CCRCC than in non-CCRCC and increased in conjunction with increased stage of tumor progression (154). Serum s-CAIX levels also correlated with the tumor size in CCRCC patients (143). In addition to serum s-CAIX, CA9 mRNA may also be a promising molecular marker of CCRCC. For example, CA9 mRNA was detected in 97% of CCRCCs and highly correlated with IHC staining of corresponding samples (155).

Similar studies have also been performed for other cancer types. For example, urine was tested from 23 patients with transitional cell carcinoma (TCC) or squamous cell carcinoma (SCC) of the urinary bladder and renal pelvis, and the presence of urine s-CAIX correlated with tumor CAIX in most cases. Additionally, s-CAIX was detected in the urine of two patients with a suspected, but unconfirmed bladder tumor, who later developed tumors within six months of testing (156). However, this correlation was not observed in TCC patients. Preoperative serum concentrations of CAIX were reported to be significantly higher in patients with high intratumoral expression of CAIX in vulvar cancer (157). Serum levels of CAIX in metastatic breast cancer and vulvar cancer were correlated with poor prognosis (157, 158). In non-small cell lung cancer, high plasma levels of s-CAIX were determined to be an independent prognostic biomarker, particularly for early-stage I or II carcinomas and were associated with significantly worse survival (111). Also, detection of s-CAIX protein by ELISA and immunocytochemistry have been used to detect malignant pleural effusions (145). The potential use of serum s-CAIX as a biomarker for therapy response was tested in patients with primary epithelial ovarian cancer, yet were not significantly altered during first-line therapy (159).

As mentioned above for CCRCC, immunohistochemical (IHC) staining may be also used to detect CAIX in tumor samples (145). IHC of CAIX is an emerging field of interest and holds a great deal of promise for quantification of disease progression, use as a predictive or prognostic indicator, and as an indicator of therapy response. Like other IHC markers, it can be used to evaluate the expression or localization of CAIX in tissue. Using standard color producing enzymes (i.e. peroxidase coupled with DAB) and amplification techniques common to the practice of IHC staining, CAIX can be visualized in a range from negative to strong positive staining intensity. CAIX is generally regarded as a specific IHC biomarker in a myriad of tissue types (117). As expected, CAIX is generally localized on the cell membrane but may also be found to a lesser degree in the cytoplasm.

6. Targeting of CAIX (imaging and therapy)

6.1 CAIX imaging

The characterization and detection of hypoxic regions within solid tumor masses is an important issue in cancer care because hypoxia is an independent predictor of poor survival in several cancer types and hypoxic cancer cells are less sensitive to chemo- and radiation- therapies (160). Therefore, a molecular imaging approach based on selective ligands to accessible proteins overexpressed at sites of hypoxia is desired. Such an agent could help physicians to decide which patients will benefit from adjunctive anti-hypoxia therapy (61, 161). Also as discussed above, since CAIX has high and broad expression among a number of aggressive and late stage types of tumors and metastases, the non-invasive imaging of CAIX expression has potential for use in cancer diagnosis and staging, and to detect and follow the response of disseminated metastatic disease to systemic and targeted therapies. Both antibodies and small MW compounds have been used for targeted imaging of CAIX expression (28).

6.1.1 CAIX specific monoclonal antibodies for imaging

G250 was characterized as a monoclonal antibody in the 1980s by Oosterwijk et al. (14), and was shown to be a very strong biomarker for CCRCC due to its absence in normal kidney tissue. The specific epitope for the G250 antibody is unknown, but it has excellent specificity for CAIX in immunohistochemical analyses. To reduce human anti-mouse antibody responses (HAMA), a chimeric version of G250 (cG250) was developed in 1997, and was recently radiolabeled with iodine-124 or zirconium-89 for diagnosis, and with iodine-131 or lutetium-177 for therapy (162, 163). Phase I of human clinical trials of positron emission tomography/computed tomography (PET/CT) imaging of CCRCC with Iodine-124 labeled cG250 have shown excellent sensitivity, specificity, positive predictive values, and negative predictive values (94,100, 100, and 90%, respectively) (164). Hence, CAIX targeted PET/CT-based molecular imaging has potential to alter the care and welfare of patients suspected of having CCRCC and could be used to monitor patients with a history of CCRCC. In addition, this approach allows for the non-invasive and non-surgical differentiation of clear cell and non-clear cell RCC. Currently, Wilex AG (Munich, Germany) is conducting phase III clinical trials using this PET imaging agent, under the name of Redectane®. Wilex is also developing a therapy application of G250 under the product name Rencarex® (165).

The M75 antibody is highly specific for CAIX (166). It recognizes the extracellular proteoglycan-like domain and is useful for western blotting, immunoprecipitation, and immunohistochemistry. A radiolabeled derivative has been developed for pre-clinical imaging of CAIX in hypoxic tumors in mouse xenograft models (167, 168) and CAIX selective accumulation of the M75 antibody labeled with iodine-125 was demonstrated in HT-29 xenografts in nude mice (167).

Fully human CAIX single-chain variable fragment (scFv) mini-antibodies, or minibodies, named A3 and CC7 were generated using phage-display technology by Ahlskog et al. (169). These minibodies recognize the extracellular carbonic anhydrase (CA) domain of human CAIX and have low nanomolar affinity to CAIX. Because they are fully human, A3 and CC7 are expected to be non-immunogenic in patients with cancer. Also, because of their smaller size, approximately a factor of 10 smaller compared to full-length antibodies, these minibodies are expected to have faster tissue distribution and clearance relative to full-sized antibodies. The A3 and CC7 minibodies do not inhibit CAIX activity and do not bind to CAXII, which shares 39% sequence identity with CAIX. We have reported that at least one of the two cell-surface markers, either CAIX or CAXII, is expressed in 100% of breast cancer axillary lymph node (ALN) metastases on our TMA.

We have also developed CAIX and CAXII specific monoclonal antibody-based near-infrared fluorescent (NIRF) molecular imaging probes (CAIXAb-680 and CAXIIAb-68) using antibodies from R&D Systems for the non-invasive detection of breast cancer ALN metastases and we have confirmed the high selectivity of the probes in vitro and in vivo using preclinical breast cancer metastasis models (152) (Figure 5).

Figure 5.

Figure 5

In vivo bioluminescence and fluorescence images of a mouse following spontaneous metastasis to the axillary lymph node from a primary mammary fat pad (MFP) xenograft tumor. MDA-mb-231 cells were used that were engineered to overexpress luciferase and constitutively express CAIX. The fluorescence image was acquired 24 h after MFP injection of CA9Ab-680 probe.

6.1.2 Imaging CAIX expression using small molecule-based probes

Carbonic anhydrase inhibitors are divided to four groups: inorganic anions, sulfonamide based compounds (sulphonamides, sulphanilamides, sulphamates and their derivatives), phenols and coumarins (1, 170172). Inorganic anions and sulfonamides inhibit CAIX by coordinating to the zinc ion within the active site, with mM to μM Ki inhibition (the anions) or μM to nM Ki inhibition (sulfonamides and their congeners) (1, 170, 173175). Phenols are generally low μM Ki CA inhibitors, and act by anchoring to the zinc-bound water molecule/hydroxide ion through a network of hydrogen bonds which stabilize the enzyme-inhibitor adduct (171). Coumarins are a novel class of CA inhibitors and act differently compared to the other inhibitors, i.e. through a non-zinc mediated mechanism. Coumarins function as suicide inhibitors, undergoing hydrolysis to 2-hydroxycinnamic acids and anchoring irreversibly at the entrance to the active site cavity (172). Although several low nanomolar and isoform-selective CAIX inhibitors based on the coumarin/thiocoumarin ring were recently reported (172), the best investigated and most robust class of inhibitors are the sulfonamides due to their high affinity, availability, and ease of chemical manipulation (1, 176). Supuran’s group has pioneered the development of specific CAIX inhibitors to target tumor growth and many sulfonamide CA inhibitors with low nM Ki inhibition have been identified in the last several years, demonstrating particular promise as potential anti-cancer agents (1, 94, 177, 178).

Interestingly, it has been demonstrated by testing several cell lines in hypoxic conditions, that sulfonamides bind to CAIX only when the enzyme is active, indicating that the active site of CAIX is only available for sulfonamide binding during hypoxia (94, 179). Hence, CAIX sulfonamides can distinguish cells that are currently in hypoxic conditions from those that were previously hypoxic, while CAIX specific antibodies do not, which is a big advantage since CAIX expression is very stable (with a half-life of ~40 h) with CAIX remaining on the cell surface for a relatively long time after reoxygenation (76, 94). Therefore, imaging with CAIX inhibitors might provide different imaging and prognostic information compared to monoclonal antibodies (98).

However, due to a high degree of homology among CA isotypes, sulfonamide-based inhibitors are generally not specific to the CAIX isoform, i.e. sulfonamides specifically bind to the catalytic sites of other CAs (1, 180). This is a problem, because unmodified sulfonamides are able to pass through the plasma membrane and can thus interact with the physiologically dominant CAI and CAII isoforms which are soluble proteins located in the cytosol or mitochondria. Several strategies are being employed to develop selective CA-isozyme inhibitors without “off-target” inhibition of intracellular CAs. These strategies include the addition of charged species or bulky entities such as FITC, albumin or hydrophilic sugar moieties, all of which prevent transportation across the plasma membrane (166). However, these strategies do not solve the problem of extracellular isoforms other than CAIX that are known to be overexpressed in cancers, e.g. CAXII. In fact, both CAIX and CAXII are transmembrane proteins with catalytic domains oriented extracellularly. Hence, the development of CAIX specific inhibitors is an area of significant interest and the design and synthesis of CAIX selective inhibitors has been recently reviewed (180, 181). For example, sulfonamides conjugated to fluorescent dye (FITC) have been developed and are shown to have membrane-impermeant properties and high affinities for CAIX. These agents have been shown to bind to cells that express CAIX in hypoxic conditions, but not in normoxic conditions or to control cells that lack CAIX expression (94). This finding suggests that hypoxia may alter the conformation of the CAIX active site, allowing access to the inhibitor. It has been proposed that interaction of the highly acidic PG domain with highly basic residues around the conserved zinc-binding histidines form a cover at the entrance to the catalytic site that open in hypoxic conditions. In support of this hypothesis, it was observed that a CAIX variant that lacks the PG domain binds to inhibitor in normoxic conditions at high levels that are comparable to binding in hypoxia (178).

In vivo studies with fluorescent sulfonamides have also shown promise for detecting CAIX expression in HT-29 and SK-RC-52 tumor xenografts (178, 182). An inhibitor-fluorescent dye conjugate was also shown to inhibit the CAIX-mediated acidification of the tumor extracellular environment (141, 179, 183). In addition, Dubois et al. reported significant accumulation of the inhibitor in the HT-29 colorectal tumors when animals were placed in hypoxic breathing conditions (7% oxygen). Furthermore, the bound CA inhibitor fraction rapidly and significantly decreased upon return to normoxic conditions and tumor reoxygenation (177).

Also very recently, a series of acetazolamide derivatives conjugated to various near-infrared fluorescent (NIRF) dyes were synthesized with the aim of imaging hypoxia-induced hCAIX expression in tumor cells in vitro, ex vivo and in vivo. The compounds demonstrated up to 50-fold selectivity for hCAIX compared to the cytosolic hCAII isoform. Some of the compounds also showed inhibition selectivity for other transmembrane isoforms as well, e.g. hCAXII and hCAXIV. A pilot in vivo study in HT-29 tumor bearing mice quantified significant accumulation (10% i.d.) in the tumor with little accumulation in other organs (except the kidneys), demonstrating the potential use of these new compounds for quantitative imaging of CAIX via fluorescence molecular tomography (FMT) (184). These results illustrate the potential of NIRF CAIX inhibitors and FMT imaging for pre-clinical studies to non-invasively quantify CAIX expression as an endogenous marker of tumor hypoxia. This capability is crucial to the study of the underlying biology of tumor hypoxia and the development and monitoring of novel anti-cancer therapies (184).

A 99mTc-labeled sulfonamide derivative has been synthesized and evaluated for visualization of CAIX expression by single photon emission computed tomography (SPECT) imaging. However despite the favorable Ki values of the tracer, a maximum tumor tracer uptake of only 0.1% ID/g was observed at 30 min post injection, indicating that the tracer is not very promising for imaging of CAIX (185). Although tracer uptake in tumors was minimal, tumor-to-blood ratios increased as a function of time and a different radionuclide with a slower decay rate may produce greater contrast at later time-points.

6.2 CAIX targeted therapy

CAIX is highly expressed in different tumor types and has relatively low expression in normal tissues; has an important role in tumor progression, acidification and metastasis; and is located on the extracellular surface of cell membranes, allowing for efficient targeting by antibodies or small molecule inhibitors. Therefore, CAIX constitutes an attractive and promising candidate marker for systemic anticancer therapy that targets disseminated metastatic disease. Two major therapeutic tools are being studied for targeting of CAIX: monoclonal antibodies and small molecule inhibitors [reviewed by (28)].

6.2.1 CAIX specific monoclonal antibodies for therapy

CAIX-specific monoclonal antibodies can be useful for anti-tumor therapy either by (a) antibody-dependent cell cytotoxicity (ADCC); (b) targeting the CAIX active site; or (c) receptor-mediated internalization for targeted delivery of various therapeutic agents, including cytotoxins and radionuclides (28).

  1. The first category of antibodies acts through ADCC. For example cG250 which targets the PG-like domain of CAIX and is marketed by WILEX AG under the trade name RENCAREX® has been extensively characterized as an anticancer immunotherapy [90]. This antibody acts via ADCC (186), and phase I and II trials have demonstrated safety and high efficacy of the antibody in either monotherapy or in combination with interferon (IFN)-α for treatment of renal cell carcinoma (RCC) (187, 188). Phase III trials have just begun using cG250 as an adjuvant therapy for reducing recurrence in surgically-treated renal cell carcinoma (RCC) patients who have a high risk of relapse and in combination with interleukin 2 (IL-2) or IFN-α for metastatic RCC (28, 187).

  2. While cG250 is directed at the PG-like domain, recent studies have focused on developing antibodies that target the catalytic domain of CAIX, thus targeting its tumorigenic functions, including pH regulation (28). For example a mAb directed against the CAIX catalytic domain has been generated using hybridoma technology and this antibody demonstrated efficient binding, internalization and persistence in cultured cells. In vivo evaluation of the antibody demonstrated an anti-cancer effect in a mouse xenograft model of colorectal carcinoma by limiting tumor growth. However, the effect was not observed when tumors were allowed to grow and become established prior to treatment (189). Though this might be the expected result, since buffer therapy has no effect on the growth of established primary cancers, while exerting a significant inhibition on the development of spontaneous metastases (104).

    Antigen-binding antibody fragments (Fab) against human CAIX have been generated by phage-display and inhibition of CAIX demonstrated in spheroid cell cultures, suggesting that these Fabs could be used to inhibit CAIX therapeutically (190). Two mAbs with high affinity to CAIX generated by phage display have also been described (169). However more studies are needed for preclinical evaluation of these antibodies.

  3. The third category of CAIX antibodies are those that are internalized and can thus be used for delivery of therapeutic payloads, such as radionuclides and cytotoxic agents. For example, stability, biodistribution and therapeutic efficacy of various radio-immunoconjugates of cG250 have been tested in nude mice with subcutaneous renal cell cancer (RCC) tumors and high therapeutic efficacy has been demonstrated by attachment of (177)Lu, (90)Y or (186)Re compared to (131)I-cG250 (191). A phase I/II trial in metastatic RCC patients is currently ongoing for (177) Lu-cG250 (192). Also in another study a CAIX mAb which was identified by phage display was conjugated to monomethyl auristatin E (a synthetic antineoplastic agent) and high efficacy as a potent anti-tumor drug confirmed in several preclinical human xenograft tumor models (193).

Alternative combination therapies are being designed that include targeting of CAIX. An example is the tyrosine kinase inhibitor sunitinib (Sutent®). This approved first line therapy for metastatic renal cell carcinoma (mRCC) has had limited clinical success. Oosterwijk et al., are developing an agent that combines Sutent® with targets of CAIX for enhancement of therapeutic effect. A second example involves treatment of the same disease (mRCC) with autologous T-cells genetically retargeted against CAIX (194).

6.2.2 CAIX targeted small molecule inhibitors for therapy

Sulfonamides are commonly used as diuretics, anti-glaucoma, anti-obesity, and anti-infective treatments. Additionally, they have potential for use as antitumor drugs and this is an active area of clinical research. In fact, with the help of X-ray crystallography, much recent progress has been made for the designing of selective CAIX inhibitors. For example, Indisulam, a sulfonamide which was recently investigated in phase II clinical trials, is considered one of the most potent anticancer sulfonamides yet and has demonstrated high anti-tumor activity in various preclinical cancer models (195). The combination of CAIX inhibitors with conventional chemotherapy may yield even better efficacy (98). Also, one of several potent bis-sulfonamide CAIX inhibitors identified by screening 1 million compounds in a DNA-encoded chemical library has demonstrated high and specific accumulation in tumor models (196). In addition, due to high-expression in hypoxia, CAIX is an attractive target for the design of hypoxia-activatable prodrugs. This may lead to a completely new approach for developing less toxic anticancer agents [see Ref (181) for review]. Recently, carbonic anhydrase inhibitors coated with gold nanoparticles, which are membrane impermeant and therefore more effective in targeting the extracellular active site of CAIX, have been developed (197). These nanoparticles exhibited more potent inhibition of CAIX compared to acetazolamide, and may be a candidate for RCC treatment or imaging and treatment of hypoxic tumors. Targeting of CAIX by small molecules in preclinical models of human cancer has been reviewed extensively in ref (7, 28).

7. CAIX and CAXII selectivity issues

CAXII is a transmembrane isoform with an extracellular active site that was originally identified because of its overexpression in renal cancer cells (43, 198). CAIX and CAXII proteins share an overall amino acid sequence identity of 35% and there is considerable homology at the catalytic site with the three key zinc-binding histidine residues conserved in both sequences (43). As mentioned above, CAIX contains an additional proteoglycan-like domain that has been implicated in cell adhesion, which is not present on CAXII (17, 20, 22). The lack of PG domains suggests that CAXII has a more neutral pH optimum for activity.

Similar to CAIX, CAXII is highly overexpressed in breast cancer (26) and other tumor types [for review see (77)], but it is also expressed in some normal tissues such as pancreas, kidney, gut and the gastrointestinal tract (199202). Several studies have shown that while CAIX expression is associated with a negative prognosis, including resistance to chemo- and radiation-therapy in breast cancer (117, 203, 204), CAXII has been associated with a favorable prognosis in invasive breast cancer (205).

Although expression of both CAIX and CAXII can be induced by hypoxia (29), CAXII is not as robustly regulated by hypoxia as CAIX and, if present, its HIF response element (HRE) has not been identified (26, 29, 54). Thus, the mechanism for CAXII induction by hypoxia is still in question. Our group has studied CAIX and CAXII expression in breast cancer tumor xenografts using multiple tumor lines. CAIX was invariably induced in regions adjacent to necrosis, while CAXII induction was generally lower compared to CAIX and that, although cell line dependent, both markers could have significant constitutive expression distal to the necrotic edge (Figure 6).

Figure 6.

Figure 6

A) CAIX and CAXII IHC staining in a region of a representative MCF-7 tumor xenograft adjacent to necrosis. B) Quantified staining intensity by distance from necrosis with pixels 0–100 being adjacent to a necrotic region.

Thus, imaging probes that specifically target CAIX may provide superior results compared to probes that non-specifically target the CA active site and can cross-react with expression of other cell-surface CA isoforms, e.g. CAXII. However in terms of therapeutic agents, the use of membrane impermeable sulfonamides, which display inhibitory constants in the low nanomolar range both towards CAIX and CAXII may be useful; because of high expression of both CAIX and CAXII is observed in several cancers (55, 152). Simultaneous silencing of both CAIX and CAXII in transfected LS174T colorectal cancer xenograft models led to substantial growth retardation that was greater compared to HIF-1α silencing alone, and greater than knockouts of CAIX or CAXII individually, which yielded substantially lower tumor growth retardation (54). Therefore drugs that can target both CAIX and CAXII may act as novel and potentially efficient drug approaches (55). However, CAXII expression in normal tissues involved in agent clearance (e.g. kidney) and toxicity (e.g. pancreas and gut) is of some concern in the context of therapy.

Specific peptides could be a promising candidate for imaging of hypoxia. Peptides present an attractive alternative to antibodies due to their small size and therefore optimal pharmacokinetic properties and high effectiveness of tumor penetration. In addition, peptides do not possess the immunogenic potential of antibodies and are easier and less expensive to produce (206). Recently, a new peptide (CaIX-P1) with specificity for the extracellular domain of CAIX was identified by phage display technology using a commercially available Ph.D.12 library and the CAIX binding affinity and biodistribution studied (207). Although the stability and affinity of CaIX-P1 was recently optimized, organ distribution studies revealed low tumor-to-blood ratios and high background distribution, which is not favorable for imaging applications (208). Therefore, further research is required for generation of peptide-based ligands with high target specificity for human carbonic anhydrase IX.

8. Conclusion

In summary, CAIX is considered to be a promising marker for targeted diagnostic imaging and systemic anticancer therapy due to its overexpression in many cancers and metastases with a limited presence in their normal tissue counterparts. CAIX expression is generally associated with a poor prognosis and is associated with tumor progression to metastasis due to its role in acidification of the extratumoral medium, which leads to both the acquisition of the metastasic phenotype and chemoresistance to weakly basic anticancer drugs.

Inhibition of CAIX activity by either specific antibodies or small molecules has been studied in the preclinical setting and more recently in clinical trials as a mode of therapy. Many classes of CAIX targeted therapeutic agents have been developed and pharmacological evaluation has demonstrated promising results. Some of these agents are now in phase III clinical trials. Concerns remain about off-target toxicities of agents that are not excluded to the extracellular environment due to interactions with intracellular CAs, and due to lack of selectivity among other extracellular CAs, e.g. CAXII which is expressed in normal tissues of concern.

There is significant interest in the development of diagnostic imaging tools for the non-invasive detection of tumor hypoxia via CAIX induced expression, for detection of CAIX expression in lymph node metastases for staging, for the diagnosis of disseminated disease and to follow response to systemic therapy during treatment of distal metastasis. In conjunction with the development of targeted therapies, there is now significant interest in the pharmaceutical industry in the development of companion targeted imaging agents for use during clinical trials. In terms of the use of CAIX as a marker for tumor hypoxia, it has been shown that CAIX is generally up-regulated in regions of hypoxia. However expression of CAIX in non-hypoxic regions of tumors has also been observed due to HIF-1 stabilization in cancer. Also, high constitutive CAXII expression in tumors and metastases may be a confounding factor for imaging agents that are not CAIX specific. However, for detection of elevated CA expression in disseminated disease, this requirement for specificity is not as important. Although, since CAXII expression is also observed in normal tissues, this could cause increased non-specific background signal in agents that are not CAIX specific.

References

  • 1.Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov. 2008;7:168–181. doi: 10.1038/nrd2467. [DOI] [PubMed] [Google Scholar]
  • 2.Supuran CT. Carbonic anhydrase inhibitors. Bioorg Med Chem Lett. 2010;20:3467–3474. doi: 10.1016/j.bmcl.2010.05.009. [DOI] [PubMed] [Google Scholar]
  • 3.Okamoto N, Fujikawa-Adachi K, Nishimori I, Taniuchi K, Onishi S. cDNA sequence of human carbonic anhydrase-related protein, CA-RP X: mRNA expressions of CA-RP X and XI in human brain. Biochim Biophys Acta. 2001;1518:311–316. doi: 10.1016/s0167-4781(01)00193-2. [DOI] [PubMed] [Google Scholar]
  • 4.Picaud SS, Muniz JR, Kramm A, Pilka ES, Kochan G, Oppermann U, Yue WW. Crystal structure of human carbonic anhydrase-related protein VIII reveals the basis for catalytic silencing. Proteins. 2009;76:507–511. doi: 10.1002/prot.22411. [DOI] [PubMed] [Google Scholar]
  • 5.Saczewski F, Innocenti A, Brzozowski Z, Slawinski J, Pomarnacka E, Kornicka A, Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors. Selective inhibition of human tumor-associated isozymes IX and XII and cytosolic isozymes I and II with some substituted-2-mercapto-benzenesulfonamides. J Enzyme Inhib Med Chem. 2006;21:563–568. doi: 10.1080/14756360600648146. [DOI] [PubMed] [Google Scholar]
  • 6.Saczewski F, Slawinski J, Kornicka A, Brzozowski Z, Pomarnacka E, Innocenti A, Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors. Inhibition of the cytosolic human isozymes I and II, and the transmembrane, tumor-associated isozymes IX and XII with substituted aromatic sulfonamides activatable in hypoxic tumors. Bioorg Med Chem Lett. 2006;16:4846–4851. doi: 10.1016/j.bmcl.2006.06.064. [DOI] [PubMed] [Google Scholar]
  • 7.Imtaiyaz Hassan M, Shajee B, Waheed A, Ahmad F, Sly WS. Structure, function and applications of carbonic anhydrase isozymes. Bioorg Med Chem. 2012;21:1570–1582. doi: 10.1016/j.bmc.2012.04.044. [DOI] [PubMed] [Google Scholar]
  • 8.Supuran CT, Scozzafava A, Casini A. Carbonic anhydrase inhibitors. Med Res Rev. 2003;23:146–189. doi: 10.1002/med.10025. [DOI] [PubMed] [Google Scholar]
  • 9.Swietach P, Wigfield S, Cobden P, Supuran CT, Harris AL, Vaughan-Jones RD. Tumor-associated carbonic anhydrase 9 spatially coordinates intracellular pH in three-dimensional multicellular growths. J Biol Chem. 2008;283:20473–20483. doi: 10.1074/jbc.M801330200. [DOI] [PubMed] [Google Scholar]
  • 10.Pastorek J, Pastorekova S, Callebaut I, Mornon JP, Zelnik V, Opavsky R, Zat’ovicova M, Liao S, Portetelle D, Stanbridge EJ. Cloning and characterization of MN, a human tumor-associated protein with a domain homologous to carbonic anhydrase and a putative helix-loop-helix DNA binding segment. Oncogene. 1994;9:2877–2888. [PubMed] [Google Scholar]
  • 11.Opavsky R, Pastorekova S, Zelnik V, Gibadulinova A, Stanbridge EJ, Zavada J, Kettmann R, Pastorek J. Human MN/CA9 gene, a novel member of the carbonic anhydrase family: structure and exon to protein domain relationships. Genomics. 1996;33:480–487. doi: 10.1006/geno.1996.0223. [DOI] [PubMed] [Google Scholar]
  • 12.Pastorekova S, Zavadova Z, Kostal M, Babusikova O, Zavada J. A novel quasi-viral agent, MaTu, is a two-component system. Virology. 1992;187:620–626. doi: 10.1016/0042-6822(92)90464-z. [DOI] [PubMed] [Google Scholar]
  • 13.Zavada J, Zavadova Z, Pastorekova S, Ciampor F, Pastorek J, Zelnik V. Expression of MaTu-MN protein in human tumor cultures and in clinical specimens. Int J Cancer. 1993;54:268–274. doi: 10.1002/ijc.2910540218. [DOI] [PubMed] [Google Scholar]
  • 14.Oosterwijk E, Ruiter DJ, Hoedemaeker PJ, Pauwels EK, Jonas U, Zwartendijk J, Warnaar SO. Monoclonal antibody G 250 recognizes a determinant present in renal-cell carcinoma and absent from normal kidney. Int J Cancer. 1986;38:489–494. doi: 10.1002/ijc.2910380406. [DOI] [PubMed] [Google Scholar]
  • 15.Grabmaier K, Vissers JL, De Weijert MC, Oosterwijk-Wakka JC, Van Bokhoven A, Brakenhoff RH, Noessner E, Mulders PA, Merkx G, Figdor CG, Adema GJ, Oosterwijk E. Molecular cloning and immunogenicity of renal cell carcinoma-associated antigen G250. Int J Cancer. 2000;85:865–870. doi: 10.1002/(sici)1097-0215(20000315)85:6<865::aid-ijc21>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 16.Hilvo M, Baranauskiene L, Salzano AM, Scaloni A, Matulis D, Innocenti A, Scozzafava A, Monti SM, Di Fiore A, De Simone G, Lindfors M, Janis J, Valjakka J, Pastorekova S, Pastorek J, Kulomaa MS, Nordlund HR, Supuran CT, Parkkila S. Biochemical characterization of CA IX, one of the most active carbonic anhydrase isozymes. J Biol Chem. 2008;283:27799–27809. doi: 10.1074/jbc.M800938200. [DOI] [PubMed] [Google Scholar]
  • 17.Zavada J, Zavadova Z, Pastorek J, Biesova Z, Jezek J, Velek J. Human tumour-associated cell adhesion protein MN/CA IX: identification of M75 epitope and of the region mediating cell adhesion. Br J Cancer. 2000;82:1808–1813. doi: 10.1054/bjoc.2000.1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.De Simone G, Supuran CT. Carbonic anhydrase IX: Biochemical and crystallographic characterization of a novel antitumor target. Biochim Biophys Acta. 2010;1804:404–409. doi: 10.1016/j.bbapap.2009.07.027. [DOI] [PubMed] [Google Scholar]
  • 19.Innocenti A, Pastorekova S, Pastorek J, Scozzafava A, De Simone G, Supuran CT. The proteoglycan region of the tumor-associated carbonic anhydrase isoform IX acts as anintrinsic buffer optimizing CO2 hydration at acidic pH values characteristic of solid tumors. Bioorg Med Chem Lett. 2009;19:5825–5828. doi: 10.1016/j.bmcl.2009.08.088. [DOI] [PubMed] [Google Scholar]
  • 20.Svastova E, Zilka N, Zat’ovicova M, Gibadulinova A, Ciampor F, Pastorek J, Pastorekova S. Carbonic anhydrase IX reduces E-cadherin-mediated adhesion of MDCK cells via interaction with beta-catenin. Exp Cell Res. 2003;290:332–345. doi: 10.1016/s0014-4827(03)00351-3. [DOI] [PubMed] [Google Scholar]
  • 21.Zavadova Z, Zavada J. Carbonic anhydrase IX (CA IX) mediates tumor cell interactions with microenvironment. Oncol Rep. 2005;13:977–982. [PubMed] [Google Scholar]
  • 22.Dorai T, Sawczuk IS, Pastorek J, Wiernik PH, Dutcher JP. The role of carbonic anhydrase IX overexpression in kidney cancer. Eur J Cancer. 2005;41:2935–2947. doi: 10.1016/j.ejca.2005.09.011. [DOI] [PubMed] [Google Scholar]
  • 23.Alterio V, Hilvo M, Di Fiore A, Supuran CT, Pan P, Parkkila S, Scaloni A, Pastorek J, Pastorekova S, Pedone C, Scozzafava A, Monti SM, De Simone G. Crystal structure of the catalytic domain of the tumor-associated human carbonic anhydrase IX. Proc Natl Acad Sci U S A. 2009;106:16233–16238. doi: 10.1073/pnas.0908301106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Whittington DA, Waheed A, Ulmasov B, Shah GN, Grubb JH, Sly WS, Christianson DW. Crystal structure of the dimeric extracellular domain of human carbonic anhydrase XII, a bitopic membrane protein overexpressed in certain cancer tumor cells. Proc Natl Acad Sci U S A. 2001;98:9545–9550. doi: 10.1073/pnas.161301298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Li Y, Wang H, Tu C, Shiverick KT, Silverman DN, Frost SC. Role of hypoxia and EGF on expression, activity, localization and phosphorylation of carbonic anhydrase IX in MDA-MB-231 breast cancer cells. Biochim Biophys Acta. 2011;1813:159–167. doi: 10.1016/j.bbamcr.2010.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ivanov S, Liao SY, Ivanova A, Danilkovitch-Miagkova A, Tarasova N, Weirich G, Merrill MJ, Proescholdt MA, Oldfield EH, Lee J, Zavada J, Waheed A, Sly W, Lerman MI, Stanbridge EJ. Expression of hypoxia-inducible cell-surface transmembrane carbonic anhydrases in human cancer. Am J Pathol. 2001;158:905–919. doi: 10.1016/S0002-9440(10)64038-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pastorekova S, Parkkila S, Parkkila AK, Opavsky R, Zelnik V, Saarnio J, Pastorek J. Carbonic anhydrase IX, MN/CA IX: analysis of stomach complementary DNA sequence and expression in human and rat alimentary tracts. Gastroenterology. 1997;112:398–408. doi: 10.1053/gast.1997.v112.pm9024293. [DOI] [PubMed] [Google Scholar]
  • 28.McDonald PC, Winum JY, Supuran CT, Dedhar S. Recent developments in targeting carbonic anhydrase IX for cancer therapeutics. Oncotarget. 2012;3:84–97. doi: 10.18632/oncotarget.422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wykoff CC, Beasley NJ, Watson PH, Turner KJ, Pastorek J, Sibtain A, Wilson GD, Turley H, Talks KL, Maxwell PH, Pugh CW, Ratcliffe PJ, Harris AL. Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res. 2000;60:7075–7083. [PubMed] [Google Scholar]
  • 30.Kaluz S, Kaluzova M, Liao SY, Lerman M, Stanbridge EJ. Transcriptional control of the tumor- and hypoxia-marker carbonic anhydrase 9: A one transcription factor (HIF-1) show? Biochim Biophys Acta. 2009;1795:162–172. doi: 10.1016/j.bbcan.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Barathova M, Takacova M, Holotnakova T, Gibadulinova A, Ohradanova A, Zatovicova M, Hulikova A, Kopacek J, Parkkila S, Supuran CT, Pastorekova S, Pastorek J. Alternative splicing variant of the hypoxia marker carbonic anhydrase IX expressed independently of hypoxia and tumour phenotype. Br J Cancer. 2008;98:129–136. doi: 10.1038/sj.bjc.6604111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zatovicova M, Sedlakova O, Svastova E, Ohradanova A, Ciampor F, Arribas J, Pastorek J, Pastorekova S. Ectodomain shedding of the hypoxia-induced carbonic anhydrase IX is a metalloprotease-dependent process regulated by TACE/ADAM17. Br J Cancer. 2005;93:1267–1276. doi: 10.1038/sj.bjc.6602861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Salceda S, Caro J. Hypoxia-inducible factor 1alpha (HIF-1alpha) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. Its stabilization by hypoxia depends on redox-induced changes. J Biol Chem. 1997;272:22642–22647. doi: 10.1074/jbc.272.36.22642. [DOI] [PubMed] [Google Scholar]
  • 34.Metzen E, Berchner-Pfannschmidt U, Stengel P, Marxsen JH, Stolze I, Klinger M, Huang WQ, Wotzlaw C, Hellwig-Burgel T, Jelkmann W, Acker H, Fandrey J. Intracellular localisation of human HIF-1 alpha hydroxylases: implications for oxygen sensing. J Cell Sci. 2003;116:1319–1326. doi: 10.1242/jcs.00318. [DOI] [PubMed] [Google Scholar]
  • 35.Semenza GL. Regulation of mammalian O2 homeostasis by hypoxia-inducible factor 1. Annu Rev Cell Dev Biol. 1999;15:551–578. doi: 10.1146/annurev.cellbio.15.1.551. [DOI] [PubMed] [Google Scholar]
  • 36.Kallio PJ, Pongratz I, Gradin K, McGuire J, Poellinger L. Activation of hypoxia-inducible factor 1alpha: posttranscriptional regulation and conformational change by recruitment of the Arnt transcription factor. Proc Natl Acad Sci U S A. 1997;94:5667–5672. doi: 10.1073/pnas.94.11.5667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271–275. doi: 10.1038/20459. [DOI] [PubMed] [Google Scholar]
  • 38.Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1) Mol Pharmacol. 2006;70:1469–1480. doi: 10.1124/mol.106.027029. [DOI] [PubMed] [Google Scholar]
  • 39.Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci. 2012;33:207–214. doi: 10.1016/j.tips.2012.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lu H, Forbes RA, Verma A. Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem. 2002;277:23111–23115. doi: 10.1074/jbc.M202487200. [DOI] [PubMed] [Google Scholar]
  • 41.Kaluzova M, Kaluz S, Lerman MI, Stanbridge EJ. DNA damage is a prerequisite for p53-mediated proteasomal degradation of HIF-1alpha in hypoxic cells and downregulation of the hypoxia marker carbonic anhydrase IX. Mol Cell Biol. 2004;24:5757–5766. doi: 10.1128/MCB.24.13.5757-5766.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kondo K, Kaelin WG., Jr The von Hippel-Lindau tumor suppressor gene. Exp Cell Res. 2001;264:117–125. doi: 10.1006/excr.2000.5139. [DOI] [PubMed] [Google Scholar]
  • 43.Ivanov SV, Kuzmin I, Wei MH, Pack S, Geil L, Johnson BE, Stanbridge EJ, Lerman MI. Down-regulation of transmembrane carbonic anhydrases in renal cell carcinoma cell lines by wild-type von Hippel-Lindau transgenes. Proc Natl Acad Sci U S A. 1998;95:12596–12601. doi: 10.1073/pnas.95.21.12596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Grabmaier K, MC AdW, Verhaegh GW, Schalken JA, Oosterwijk E. Strict regulation of CAIX(G250/MN) by HIF-1alpha in clear cell renal cell carcinoma. Oncogene. 2004;23:5624–5631. doi: 10.1038/sj.onc.1207764. [DOI] [PubMed] [Google Scholar]
  • 45.Das B, Tsuchida R, Malkin D, Koren G, Baruchel S, Yeger H. Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction. Stem Cells. 2008;26:1818–1830. doi: 10.1634/stemcells.2007-0724. [DOI] [PubMed] [Google Scholar]
  • 46.Mohyeldin A, Garzon-Muvdi T, Quinones-Hinojosa A. Oxygen in stem cell biology: a critical component of the stem cell niche. Cell Stem Cell. 2010;7:150–161. doi: 10.1016/j.stem.2010.07.007. [DOI] [PubMed] [Google Scholar]
  • 47.Xing F, Okuda H, Watabe M, Kobayashi A, Pai SK, Liu W, Pandey PR, Fukuda K, Hirota S, Sugai T, Wakabayshi G, Koeda K, Kashiwaba M, Suzuki K, Chiba T, Endo M, Mo YY, Watabe K. Hypoxia-induced Jagged2 promotes breast cancer metastasis and self-renewal of cancer stem-like cells. Oncogene. 2011;30:4075–4086. doi: 10.1038/onc.2011.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lock FE, McDonald PC, Lou Y, Serrano I, Chafe SC, Ostlund C, Aparicio S, Winum JY, Supuran CT, Dedhar S. Targeting carbonic anhydrase IX depletes breast cancer stem cells within the hypoxic niche. Oncogene. 2012:1–10. doi: 10.1038/onc.2012.550. [DOI] [PubMed] [Google Scholar]
  • 49.Horree N, van Diest PJ, Sie-Go DM, Heintz AP. The invasive front in endometrial carcinoma: higher proliferation and associated derailment of cell cycle regulators. Hum Pathol. 2007;38:1232–1238. doi: 10.1016/j.humpath.2007.01.008. [DOI] [PubMed] [Google Scholar]
  • 50.Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner T. Opinion: migrating cancer stem cells - an integrated concept of malignant tumour progression. Nat Rev Cancer. 2005;5:744–749. doi: 10.1038/nrc1694. [DOI] [PubMed] [Google Scholar]
  • 51.Gut MO, Parkkila S, Vernerova Z, Rohde E, Zavada J, Hocker M, Pastorek J, Karttunen T, Gibadulinova A, Zavadova Z, Knobeloch KP, Wiedenmann B, Svoboda J, Horak I, Pastorekova S. Gastric hyperplasia in mice with targeted disruption of the carbonic anhydrase gene Car9. Gastroenterology. 2002;123:1889–1903. doi: 10.1053/gast.2002.37052. [DOI] [PubMed] [Google Scholar]
  • 52.Leppilampi M, Karttunen TJ, Kivela J, Gut MO, Pastorekova S, Pastorek J, Parkkila S. Gastric pit cell hyperplasia and glandular atrophy in carbonic anhydrase IX knockout mice: studies on two strains C57/BL6 and BALB/C. Transgenic Res. 2005;14:655–663. doi: 10.1007/s11248-005-7215-z. [DOI] [PubMed] [Google Scholar]
  • 53.Pan P, Leppilampi M, Pastorekova S, Pastorek J, Waheed A, Sly WS, Parkkila S. Carbonic anhydrase gene expression in CA II-deficient (Car2-/-) and CA IX-deficient (Car9-/-) mice. J Physiol. 2006;571:319–327. doi: 10.1113/jphysiol.2005.102590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chiche J, Ilc K, Laferriere J, Trottier E, Dayan F, Mazure NM, Brahimi-Horn MC, Pouyssegur J. Hypoxia-inducible carbonic anhydrase IX and XII promote tumor cell growth by counteracting acidosis through the regulation of the intracellular pH. Cancer Res. 2009;69:358–368. doi: 10.1158/0008-5472.CAN-08-2470. [DOI] [PubMed] [Google Scholar]
  • 55.Chiche J, Ilc K, Brahimi-Horn MC, Pouyssegur J. Membrane-bound carbonic anhydrases are key pH regulators controlling tumor growth and cell migration. Adv Enzyme Regul. 2010;50:20–33. doi: 10.1016/j.advenzreg.2009.10.005. [DOI] [PubMed] [Google Scholar]
  • 56.Harris AL. Hypoxia--a key regulatory factor in tumour growth. Nat Rev Cancer. 2002;2:38–47. doi: 10.1038/nrc704. [DOI] [PubMed] [Google Scholar]
  • 57.Wouters A, Pauwels B, Lardon F, Vermorken JB. Review: implications of in vitro research on the effect of radiotherapy and chemotherapy under hypoxic conditions. Oncologist. 2007;12:690–712. doi: 10.1634/theoncologist.12-6-690. [DOI] [PubMed] [Google Scholar]
  • 58.Comerford KM, Wallace TJ, Karhausen J, Louis NA, Montalto MC, Colgan SP. Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res. 2002;62:3387–3394. [PubMed] [Google Scholar]
  • 59.Rohwer N, Cramer T. Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist Updat. 2011;14:191–201. doi: 10.1016/j.drup.2011.03.001. [DOI] [PubMed] [Google Scholar]
  • 60.Hockel M, Schlenger K, Mitze M, Schaffer U, Vaupel P. Hypoxia and Radiation Response in Human Tumors. Semin Radiat Oncol. 1996;6:3–9. doi: 10.1053/SRAO0060003. [DOI] [PubMed] [Google Scholar]
  • 61.Evans SM, Koch CJ. Prognostic significance of tumor oxygenation in humans. Cancer Lett. 2003;195:1–16. doi: 10.1016/s0304-3835(03)00012-0. [DOI] [PubMed] [Google Scholar]
  • 62.Sun JD, Liu Q, Wang J, Ahluwalia D, Ferraro D, Wang Y, Duan JX, Ammons WS, Curd JG, Matteucci MD, Hart CP. Selective tumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibits tumor growth in preclinical models of cancer. Clin Cancer Res. 2012;18:758–770. doi: 10.1158/1078-0432.CCR-11-1980. [DOI] [PubMed] [Google Scholar]
  • 63.Gatenby RA, Kessler HB, Rosenblum JS, Coia LR, Moldofsky PJ, Hartz WH, Broder GJ. Oxygen distribution in squamous cell carcinoma metastases and its relationship to outcome of radiation therapy. Int J Radiat Oncol Biol Phys. 1988;14:831–838. doi: 10.1016/0360-3016(88)90002-8. [DOI] [PubMed] [Google Scholar]
  • 64.Mees G, Dierckx R, Vangestel C, Van de Wiele C. Molecular imaging of hypoxia with radiolabelled agents. Eur J Nucl Med Mol Imaging. 2009;36:1674–1686. doi: 10.1007/s00259-009-1195-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nozue M, Lee I, Yuan F, Teicher BA, Brizel DM, Dewhirst MW, Milross CG, Milas L, Song CW, Thomas CD, Guichard M, Evans SM, Koch CJ, Lord EM, Jain RK, Suit HD. Interlaboratory variation in oxygen tension measurement by Eppendorf “Histograph” and comparison with hypoxic marker. J Surg Oncol. 1997;66:30–38. doi: 10.1002/(sici)1096-9098(199709)66:1<30::aid-jso7>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 66.Jenkins WT, Evans SM, Koch CJ. Hypoxia and necrosis in rat 9L glioma and Morris 7777 hepatoma tumors: comparative measurements using EF5 binding and the Eppendorf needle electrode. Int J Radiat Oncol Biol Phys. 2000;46:1005–1017. doi: 10.1016/s0360-3016(99)00342-9. [DOI] [PubMed] [Google Scholar]
  • 67.Cook GJ. Oncological molecular imaging: nuclear medicine techniques. Br J Radiol. 2003;76:S152–158. doi: 10.1259/bjr/16098061. [DOI] [PubMed] [Google Scholar]
  • 68.Hoskin PJ, Sibtain A, Daley FM, Wilson GD. GLUT1 and CAIX as intrinsic markers of hypoxia in bladder cancer: relationship with vascularity and proliferation as predictors of outcome of ARCON. Br J Cancer. 2003;89:1290–1297. doi: 10.1038/sj.bjc.6601260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Jankovic B, Aquino-Parsons C, Raleigh JA, Stanbridge EJ, Durand RE, Banath JP, MacPhail SH, Olive PL. Comparison between pimonidazole binding, oxygen electrode measurements, and expression of endogenous hypoxia markers in cancer of the uterine cervix. Cytometry B Clin Cytom. 2006;70:45–55. doi: 10.1002/cyto.b.20086. [DOI] [PubMed] [Google Scholar]
  • 70.Li XF, Carlin S, Urano M, Russell J, Ling CC, O’Donoghue JA. Visualization of hypoxia in microscopic tumors by immunofluorescent microscopy. Cancer Res. 2007;67:7646–7653. doi: 10.1158/0008-5472.CAN-06-4353. [DOI] [PubMed] [Google Scholar]
  • 71.Olive PL, Aquino-Parsons C, MacPhail SH, Liao SY, Raleigh JA, Lerman MI, Stanbridge EJ. Carbonic anhydrase 9 as an endogenous marker for hypoxic cells in cervical cancer. Cancer Res. 2001;61:8924–8929. [PubMed] [Google Scholar]
  • 72.Sobhanifar S, Aquino-Parsons C, Stanbridge EJ, Olive P. Reduced expression of hypoxia-inducible factor-1alpha in perinecrotic regions of solid tumors. Cancer Res. 2005;65:7259–7266. doi: 10.1158/0008-5472.CAN-04-4480. [DOI] [PubMed] [Google Scholar]
  • 73.Carlin S, Khan N, Ku T, Longo VA, Larson SM, Smith-Jones PM. Molecular targeting of carbonic anhydrase IX in mice with hypoxic HT29 colorectal tumor xenografts. PLoS One. 2010;5:e10857. doi: 10.1371/journal.pone.0010857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Beasley NJ, Wykoff CC, Watson PH, Leek R, Turley H, Gatter K, Pastorek J, Cox GJ, Ratcliffe P, Harris AL. Carbonic anhydrase IX, an endogenous hypoxia marker, expression in head and neck squamous cell carcinoma and its relationship to hypoxia, necrosis, and microvessel density. Cancer Res. 2001;61:5262–5267. [PubMed] [Google Scholar]
  • 75.Mayer A, Hockel M, Vaupel P. Carbonic anhydrase IX expression and tumor oxygenation status do not correlate at the microregional level in locally advanced cancers of the uterine cervix. Clin Cancer Res. 2005;11:7220–7225. doi: 10.1158/1078-0432.CCR-05-0869. [DOI] [PubMed] [Google Scholar]
  • 76.Rafajova M, Zatovicova M, Kettmann R, Pastorek J, Pastorekova S. Induction by hypoxia combined with low glucose or low bicarbonate and high posttranslational stability upon reoxygenation contribute to carbonic anhydrase IX expression in cancer cells. Int J Oncol. 2004;24:995–1004. [PubMed] [Google Scholar]
  • 77.Chiche J, Brahimi-Horn MC, Pouyssegur J. Tumour hypoxia induces a metabolic shift causing acidosis: a common feature in cancer. J Cell Mol Med. 2010;14:771–794. doi: 10.1111/j.1582-4934.2009.00994.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brown JM, Wilson WR. Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer. 2004;4:437–447. doi: 10.1038/nrc1367. [DOI] [PubMed] [Google Scholar]
  • 79.Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat Rev Cancer. 2008;8:425–437. doi: 10.1038/nrc2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Koritzinsky M, Magagnin MG, van den Beucken T, Seigneuric R, Savelkouls K, Dostie J, Pyronnet S, Kaufman RJ, Weppler SA, Voncken JW, Lambin P, Koumenis C, Sonenberg N, Wouters BG. Gene expression during acute and prolonged hypoxia is regulated by distinct mechanisms of translational control. EMBO J. 2006;25:1114–1125. doi: 10.1038/sj.emboj.7600998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Brahimi-Horn MC, Chiche J, Pouyssegur J. Hypoxia and cancer. J Mol Med (Berl) 2007;85:1301–1307. doi: 10.1007/s00109-007-0281-3. [DOI] [PubMed] [Google Scholar]
  • 82.Brahimi-Horn MC, Chiche J, Pouyssegur J. Hypoxia signalling controls metabolic demand. Curr Opin Cell Biol. 2007;19:223–229. doi: 10.1016/j.ceb.2007.02.003. [DOI] [PubMed] [Google Scholar]
  • 83.Brahimi-Horn MC, Pouyssegur J. Oxygen, a source of life and stress. FEBS Lett. 2007;581:3582–3591. doi: 10.1016/j.febslet.2007.06.018. [DOI] [PubMed] [Google Scholar]
  • 84.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 85.Chambard JC, Pouyssegur J. Intracellular pH controls growth factor-induced ribosomal protein S6 phosphorylation and protein synthesis in the G0----G1 transition of fibroblasts. Exp Cell Res. 1986;164:282–294. doi: 10.1016/0014-4827(86)90029-7. [DOI] [PubMed] [Google Scholar]
  • 86.Pouyssegur J, Sardet C, Franchi A, L’Allemain G, Paris S. A specific mutation abolishing Na+/H+ antiport activity in hamster fibroblasts precludes growth at neutral and acidic pH. Proc Natl Acad Sci U S A. 1984;81:4833–4837. doi: 10.1073/pnas.81.15.4833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Roos A, Boron WF. Intracellular pH. Physiol Rev. 1981;61:296–434. doi: 10.1152/physrev.1981.61.2.296. [DOI] [PubMed] [Google Scholar]
  • 88.Neri D, Supuran CT. Interfering with pH regulation in tumours as a therapeutic strategy. Nat Rev Drug Discov. 2011;10:767–777. doi: 10.1038/nrd3554. [DOI] [PubMed] [Google Scholar]
  • 89.Parks SK, Chiche J, Pouyssegur J. pH control mechanisms of tumor survival and growth. J Cell Physiol. 2011;226:299–308. doi: 10.1002/jcp.22400. [DOI] [PubMed] [Google Scholar]
  • 90.Cardone RA, Casavola V, Reshkin SJ. The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis. Nat Rev Cancer. 2005;5:786–795. doi: 10.1038/nrc1713. [DOI] [PubMed] [Google Scholar]
  • 91.Counillon L, Pouyssegur J. The expanding family of eucaryotic Na(+)/H(+) exchangers. J Biol Chem. 2000;275:1–4. doi: 10.1074/jbc.275.1.1. [DOI] [PubMed] [Google Scholar]
  • 92.Ullah MS, Davies AJ, Halestrap AP. The plasma membrane lactate transporter MCT4, but not MCT1, is up-regulated by hypoxia through a HIF-1alpha-dependent mechanism. J Biol Chem. 2006;281:9030–9037. doi: 10.1074/jbc.M511397200. [DOI] [PubMed] [Google Scholar]
  • 93.Karumanchi SA, Jiang L, Knebelmann B, Stuart-Tilley AK, Alper SL, Sukhatme VP. VHL tumor suppressor regulates Cl-/HCO3- exchange and Na+/H+ exchange activities in renal carcinoma cells. Physiol Genomics. 2001;5:119–128. doi: 10.1152/physiolgenomics.2001.5.3.119. [DOI] [PubMed] [Google Scholar]
  • 94.Svastova E, Hulikova A, Rafajova M, Zat’ovicova M, Gibadulinova A, Casini A, Cecchi A, Scozzafava A, Supuran CT, Pastorek J, Pastorekova S. Hypoxia activates the capacity of tumor-associated carbonic anhydrase IX to acidify extracellular pH. FEBS Lett. 2004;577:439–445. doi: 10.1016/j.febslet.2004.10.043. [DOI] [PubMed] [Google Scholar]
  • 95.Raghunand N, Gatenby RA, Gillies RJ. Microenvironmental and cellular consequences of altered blood flow in tumours. Br J Radiol. 2003;76:S11–22. doi: 10.1259/bjr/12913493. [DOI] [PubMed] [Google Scholar]
  • 96.Li Y, Tu C, Wang H, Silverman DN, Frost SC. Catalysis and pH control by membrane-associated carbonic anhydrase IX in MDA-MB-231 breast cancer cells. J Biol Chem. 2011;286:15789–15796. doi: 10.1074/jbc.M110.188524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Swietach P, Vaughan-Jones RD, Harris AL. Regulation of tumor pH and the role of carbonic anhydrase 9. Cancer Metastasis Rev. 2007;26:299–310. doi: 10.1007/s10555-007-9064-0. [DOI] [PubMed] [Google Scholar]
  • 98.Pastorekova S, Ratcliffe PJ, Pastorek J. Molecular mechanisms of carbonic anhydrase IX-mediated pH regulation under hypoxia. BJU Int. 2008;101(Suppl 4):8–15. doi: 10.1111/j.1464-410X.2008.07642.x. [DOI] [PubMed] [Google Scholar]
  • 99.Wojtkowiak JW, Verduzco D, Schramm KJ, Gillies RJ. Drug resistance and cellular adaptation to tumor acidic pH microenvironment. Mol Pharm. 2011;8:2032–2038. doi: 10.1021/mp200292c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.De Milito A, Canese R, Marino ML, Borghi M, Iero M, Villa A, Venturi G, Lozupone F, Iessi E, Logozzi M, Della Mina P, Santinami M, Rodolfo M, Podo F, Rivoltini L, Fais S. pH-dependent antitumor activity of proton pump inhibitors against human melanoma is mediated by inhibition of tumor acidity. Int J Cancer. 2010;127:207–219. doi: 10.1002/ijc.25009. [DOI] [PubMed] [Google Scholar]
  • 101.Swietach P, Patiar S, Supuran CT, Harris AL, Vaughan-Jones RD. The role of carbonic anhydrase 9 in regulating extracellular and intracellular ph in three-dimensional tumor cell growths. J Biol Chem. 2009;284:20299–20310. doi: 10.1074/jbc.M109.006478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Swietach P, Hulikova A, Vaughan-Jones RD, Harris AL. New insights into the physiological role of carbonic anhydrase IX in tumour pH regulation. Oncogene. 2010;29:6509–6521. doi: 10.1038/onc.2010.455. [DOI] [PubMed] [Google Scholar]
  • 103.Raghunand N, He X, van Sluis R, Mahoney B, Baggett B, Taylor CW, Paine-Murrieta G, Roe D, Bhujwalla ZM, Gillies RJ. Enhancement of chemotherapy by manipulation of tumour pH. Br J Cancer. 1999;80:1005–1011. doi: 10.1038/sj.bjc.6690455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Robey IF, Baggett BK, Kirkpatrick ND, Roe DJ, Dosescu J, Sloane BF, Hashim AI, Morse DL, Raghunand N, Gatenby RA, Gillies RJ. Bicarbonate increases tumor pH and inhibits spontaneous metastases. Cancer Res. 2009;69:2260–2268. doi: 10.1158/0008-5472.CAN-07-5575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ibrahim-Hashim A, Cornnell HH, Abrahams D, Lloyd M, Bui M, Gillies RJ, Gatenby RA. Systemic buffers inhibit carcinogenesis in TRAMP mice. J Urol. 2012;188:624–631. doi: 10.1016/j.juro.2012.03.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Silva AS, Yunes JA, Gillies RJ, Gatenby RA. The potential role of systemic buffers in reducing intratumoral extracellular pH and acid-mediated invasion. Cancer Res. 2009;69:2677–2684. doi: 10.1158/0008-5472.CAN-08-2394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Gullotti E, Yeo Y. Extracellularly activated nanocarriers: a new paradigm of tumor targeted drug delivery. Mol Pharm. 2009;6:1041–1051. doi: 10.1021/mp900090z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Rios-Doria J, Carie A, Costich T, Burke B, Skaff H, Panicucci R, Sill K. A versatile polymer micelle drug delivery system for encapsulation and in vivo stabilization of hydrophobic anticancer drugs. J Drug Deliv. 2012;2012:951741. doi: 10.1155/2012/951741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Swinson DE, Jones JL, Richardson D, Wykoff C, Turley H, Pastorek J, Taub N, Harris AL, O’Byrne KJ. Carbonic anhydrase IX expression, a novel surrogate marker of tumor hypoxia, is associated with a poor prognosis in non-small-cell lung cancer. J Clin Oncol. 2003;21:473–482. doi: 10.1200/JCO.2003.11.132. [DOI] [PubMed] [Google Scholar]
  • 110.Kim SJ, Rabbani ZN, Dewhirst MW, Vujaskovic Z, Vollmer RT, Schreiber EG, Oosterwijk E, Kelley MJ. Expression of HIF-1alpha, CA IX, VEGF, and MMP-9 in surgically resected non-small cell lung cancer. Lung Cancer. 2005;49:325–335. doi: 10.1016/j.lungcan.2005.03.036. [DOI] [PubMed] [Google Scholar]
  • 111.Ilie M, Mazure NM, Hofman V, Ammadi RE, Ortholan C, Bonnetaud C, Havet K, Venissac N, Mograbi B, Mouroux J, Pouyssegur J, Hofman P. High levels of carbonic anhydrase IX in tumour tissue and plasma are biomarkers of poor prognostic in patients with non-small cell lung cancer. Br J Cancer. 2010;102:1627–1635. doi: 10.1038/sj.bjc.6605690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Saarnio J, Parkkila S, Parkkila AK, Haukipuro K, Pastorekova S, Pastorek J, Kairaluoma MI, Karttunen TJ. Immunohistochemical study of colorectal tumors for expression of a novel transmembrane carbonic anhydrase, MN/CA IX, with potential value as a marker of cell proliferation. Am J Pathol. 1998;153:279–285. doi: 10.1016/S0002-9440(10)65569-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Korkeila E, Talvinen K, Jaakkola PM, Minn H, Syrjanen K, Sundstrom J, Pyrhonen S. Expression of carbonic anhydrase IX suggests poor outcome in rectal cancer. Br J Cancer. 2009;100:874–880. doi: 10.1038/sj.bjc.6604949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Niemela AM, Hynninen P, Mecklin JP, Kuopio T, Kokko A, Aaltonen L, Parkkila AK, Pastorekova S, Pastorek J, Waheed A, Sly WS, Orntoft TF, Kruhoffer M, Haapasalo H, Parkkila S, Kivela AJ. Carbonic anhydrase IX is highly expressed in hereditary nonpolyposis colorectal cancer. Cancer Epidemiol Biomarkers Prev. 2007;16:1760–1766. doi: 10.1158/1055-9965.EPI-07-0080. [DOI] [PubMed] [Google Scholar]
  • 115.Chen J, Rocken C, Hoffmann J, Kruger S, Lendeckel U, Rocco A, Pastorekova S, Malfertheiner P, Ebert MP. Expression of carbonic anhydrase 9 at the invasion front of gastric cancers. Gut. 2005;54:920–927. doi: 10.1136/gut.2004.047340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Juhasz M, Chen J, Lendeckel U, Kellner U, Kasper HU, Tulassay Z, Pastorekova S, Malfertheiner P, Ebert MP. Expression of carbonic anhydrase IX in human pancreatic cancer. Aliment Pharmacol Ther. 2003;18:837–846. doi: 10.1046/j.1365-2036.2003.01738.x. [DOI] [PubMed] [Google Scholar]
  • 117.Chia SK, Wykoff CC, Watson PH, Han C, Leek RD, Pastorek J, Gatter KC, Ratcliffe P, Harris AL. Prognostic significance of a novel hypoxia-regulated marker, carbonic anhydrase IX, in invasive breast carcinoma. J Clin Oncol. 2001;19:3660–3668. doi: 10.1200/JCO.2001.19.16.3660. [DOI] [PubMed] [Google Scholar]
  • 118.Trastour C, Benizri E, Ettore F, Ramaioli A, Chamorey E, Pouyssegur J, Berra E. HIF-1alpha and CA IX staining in invasive breast carcinomas: prognosis and treatment outcome. Int J Cancer. 2007;120:1451–1458. doi: 10.1002/ijc.22436. [DOI] [PubMed] [Google Scholar]
  • 119.Tan EY, Yan M, Campo L, Han C, Takano E, Turley H, Candiloro I, Pezzella F, Gatter KC, Millar EK, O’Toole SA, McNeil CM, Crea P, Segara D, Sutherland RL, Harris AL, Fox SB. The key hypoxia regulated gene CAIX is upregulated in basal-like breast tumours and is associated with resistance to chemotherapy. Br J Cancer. 2009;100:405–411. doi: 10.1038/sj.bjc.6604844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Hussain SA, Ganesan R, Reynolds G, Gross L, Stevens A, Pastorek J, Murray PG, Perunovic B, Anwar MS, Billingham L, James ND, Spooner D, Poole CJ, Rea DW, Palmer DH. Hypoxia-regulated carbonic anhydrase IX expression is associated with poor survival in patients with invasive breast cancer. Br J Cancer. 2007;96:104–109. doi: 10.1038/sj.bjc.6603530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Neumeister VM, Sullivan CA, Lindner R, Lezon-Geyda K, Li J, Zavada J, Martel M, Glazer PM, Tuck DP, Rimm DL, Harris L. Hypoxia-induced protein CAIX is associated with somatic loss of BRCA1 protein and pathway activity in triple negative breast cancer. Breast Cancer Res Treat. 2012;136:67–75. doi: 10.1007/s10549-012-2232-0. [DOI] [PubMed] [Google Scholar]
  • 122.Loncaster JA, Harris AL, Davidson SE, Logue JP, Hunter RD, Wycoff CC, Pastorek J, Ratcliffe PJ, Stratford IJ, West CM. Carbonic anhydrase (CA IX) expression, a potential new intrinsic marker of hypoxia: correlations with tumor oxygen measurements and prognosis in locally advanced carcinoma of the cervix. Cancer Res. 2001;61:6394–6399. [PubMed] [Google Scholar]
  • 123.Kim JY, Shin HJ, Kim TH, Cho KH, Shin KH, Kim BK, Roh JW, Lee S, Park SY, Hwang YJ, Han IO. Tumor-associated carbonic anhydrases are linked to metastases in primary cervical cancer. J Cancer Res Clin Oncol. 2006;132:302–308. doi: 10.1007/s00432-005-0068-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Lee S, Shin HJ, Han IO, Hong EK, Park SY, Roh JW, Shin KH, Kim TH, Kim JY. Tumor carbonic anhydrase 9 expression is associated with the presence of lymph node metastases in uterine cervical cancer. Cancer Sci. 2007;98:329–333. doi: 10.1111/j.1349-7006.2007.00396.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Woelber L, Kress K, Kersten JF, Choschzick M, Kilic E, Herwig U, Lindner C, Schwarz J, Jaenicke F, Mahner S, Milde-Langosch K, Mueller V, Ihnen M. Carbonic anhydrase IX in tumor tissue and sera of patients with primary cervical cancer. BMC Cancer. 2011;11:12. doi: 10.1186/1471-2407-11-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Klatte T, Seligson DB, Rao JY, Yu H, de Martino M, Kawaoka K, Wong SG, Belldegrun AS, Pantuck AJ. Carbonic anhydrase IX in bladder cancer: a diagnostic, prognostic, and therapeutic molecular marker. Cancer. 2009;115:1448–1458. doi: 10.1002/cncr.24163. [DOI] [PubMed] [Google Scholar]
  • 127.Choschzick M, Oosterwijk E, Muller V, Woelber L, Simon R, Moch H, Tennstedt P. Overexpression of carbonic anhydrase IX (CAIX) is an independent unfavorable prognostic marker in endometrioid ovarian cancer. Virchows Arch. 2011;459:193–200. doi: 10.1007/s00428-011-1105-y. [DOI] [PubMed] [Google Scholar]
  • 128.Nordfors K, Haapasalo J, Korja M, Niemela A, Laine J, Parkkila AK, Pastorekova S, Pastorek J, Waheed A, Sly WS, Parkkila S, Haapasalo H. The tumour-associated carbonic anhydrases CA II, CA IX and CA XII in a group of medulloblastomas and supratentorial primitive neuroectodermal tumours: an association of CA IX with poor prognosis. BMC Cancer. 2010;10:148. doi: 10.1186/1471-2407-10-148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Proescholdt MA, Merrill MJ, Stoerr EM, Lohmeier A, Pohl F, Brawanski A. Function of carbonic anhydrase IX in glioblastoma multiforme. Neuro Oncol. 2012;14:1357–1366. doi: 10.1093/neuonc/nos216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Hoogsteen IJ, Marres HA, Wijffels KI, Rijken PF, Peters JP, van den Hoogen FJ, Oosterwijk E, van der Kogel AJ, Kaanders JH. Colocalization of carbonic anhydrase 9 expression and cell proliferation in human head and neck squamous cell carcinoma. Clin Cancer Res. 2005;11:97–106. [PubMed] [Google Scholar]
  • 131.De Schutter H, Landuyt W, Verbeken E, Goethals L, Hermans R, Nuyts S. The prognostic value of the hypoxia markers CA IX and GLUT 1 and the cytokines VEGF and IL 6 in head and neck squamous cell carcinoma treated by radiotherapy +/- chemotherapy. BMC Cancer. 2005;5:42. doi: 10.1186/1471-2407-5-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Koukourakis MI, Bentzen SM, Giatromanolaki A, Wilson GD, Daley FM, Saunders MI, Dische S, Sivridis E, Harris AL. Endogenous markers of two separate hypoxia response pathways (hypoxia inducible factor 2 alpha and carbonic anhydrase 9) are associated with radiotherapy failure in head and neck cancer patients recruited in the CHART randomized trial. J Clin Oncol. 2006;24:727–735. doi: 10.1200/JCO.2005.02.7474. [DOI] [PubMed] [Google Scholar]
  • 133.Haapasalo JA, Nordfors KM, Hilvo M, Rantala IJ, Soini Y, Parkkila AK, Pastorekova S, Pastorek J, Parkkila SM, Haapasalo HK. Expression of carbonic anhydrase IX in astrocytic tumors predicts poor prognosis. Clin Cancer Res. 2006;12:473–477. doi: 10.1158/1078-0432.CCR-05-0848. [DOI] [PubMed] [Google Scholar]
  • 134.Eckert AW, Kappler M, Schubert J, Taubert H. Correlation of expression of hypoxia-related proteins with prognosis in oral squamous cell carcinoma patients. Oral Maxillofac Surg. 2012;16:189–196. doi: 10.1007/s10006-012-0335-8. [DOI] [PubMed] [Google Scholar]
  • 135.Eckert AW, Lautner MH, Schutze A, Bolte K, Bache M, Kappler M, Schubert J, Taubert H, Bilkenroth U. Co-expression of Hif1alpha and CAIX is associated with poor prognosis in oral squamous cell carcinoma patients. J Oral Pathol Med. 2010;39:313–317. doi: 10.1111/j.1600-0714.2009.00829.x. [DOI] [PubMed] [Google Scholar]
  • 136.Choi SW, Kim JY, Park JY, Cha IH, Kim J, Lee S. Expression of carbonic anhydrase IX is associated with postoperative recurrence and poor prognosis in surgically treated oral squamous cell carcinoma. Hum Pathol. 2008;39:1317–1322. doi: 10.1016/j.humpath.2007.10.026. [DOI] [PubMed] [Google Scholar]
  • 137.Pouyssegur J, Dayan F, Mazure NM. Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature. 2006;441:437–443. doi: 10.1038/nature04871. [DOI] [PubMed] [Google Scholar]
  • 138.Liao SY, Aurelio ON, Jan K, Zavada J, Stanbridge EJ. Identification of the MN/CA9 protein as a reliable diagnostic biomarker of clear cell carcinoma of the kidney. Cancer Res. 1997;57:2827–2831. [PubMed] [Google Scholar]
  • 139.Muriel Lopez C, Esteban E, Berros JP, Pardo P, Astudillo A, Izquierdo M, Crespo G, Sanmamed M, Fonseca PJ, Martinez-Camblor P. Prognostic factors in patients with advanced renal cell carcinoma. Clin Genitourin Cancer. 2012;10:262–270. doi: 10.1016/j.clgc.2012.06.005. [DOI] [PubMed] [Google Scholar]
  • 140.Jiang YD, Ren F, Zheng SB. Value of MN/CAIX in the diagnosis of renal cell carcinoma. Nan Fang Yi Ke Da Xue Xue Bao. 2012;32:412–414. [PubMed] [Google Scholar]
  • 141.Lou Y, McDonald PC, Oloumi A, Chia S, Ostlund C, Ahmadi A, Kyle A, Auf dem Keller U, Leung S, Huntsman D, Clarke B, Sutherland BW, Waterhouse D, Bally M, Roskelley C, Overall CM, Minchinton A, Pacchiano F, Carta F, Scozzafava A, Touisni N, Winum JY, Supuran CT, Dedhar S. Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors. Cancer Res. 2011;71:3364–3376. doi: 10.1158/0008-5472.CAN-10-4261. [DOI] [PubMed] [Google Scholar]
  • 142.Choueiri TK, Regan MM, Rosenberg JE, Oh WK, Clement J, Amato AM, McDermott D, Cho DC, Atkins MB, Signoretti S. Carbonic anhydrase IX and pathological features as predictors of outcome in patients with metastatic clear-cell renal cell carcinoma receiving vascular endothelial growth factor-targeted therapy. BJU Int. 2010;106:772–778. doi: 10.1111/j.1464-410X.2010.09218.x. [DOI] [PubMed] [Google Scholar]
  • 143.Zhou GX, Ireland J, Rayman P, Finke J, Zhou M. Quantification of carbonic anhydrase IX expression in serum and tissue of renal cell carcinoma patients using enzyme-linked immunosorbent assay: prognostic and diagnostic potentials. Urology. 2010;75:257–261. doi: 10.1016/j.urology.2009.09.052. [DOI] [PubMed] [Google Scholar]
  • 144.Eriksen JG, Overgaard J. Lack of prognostic and predictive value of CA IX in radiotherapy of squamous cell carcinoma of the head and neck with known modifiable hypoxia: an evaluation of the DAHANCA 5 study. Radiother Oncol. 2007;83:383–388. doi: 10.1016/j.radonc.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 145.Liao ND, Lee WY. Detection of carbonic anhydrase IX protein in the diagnosis of malignant pleural effusion by enzyme-linked immunosorbent assay and immunocytochemistry. Cancer Cytopathol. 2012;120:269–275. doi: 10.1002/cncy.21191. [DOI] [PubMed] [Google Scholar]
  • 146.Garcia S, Dales JP, Charafe-Jauffret E, Carpentier-Meunier S, Andrac-Meyer L, Jacquemier J, Andonian C, Lavaut MN, Allasia C, Bonnier P, Charpin C. Poor prognosis in breast carcinomas correlates with increased expression of targetable CD146 and c-Met and with proteomic basal-like phenotype. Hum Pathol. 2007;38:830–841. doi: 10.1016/j.humpath.2006.11.015. [DOI] [PubMed] [Google Scholar]
  • 147.Liao SY, Darcy KM, Randall LM, Tian C, Monk BJ, Burger RA, Fruehauf JP, Peters WA, Stock RJ, Stanbridge EJ. Prognostic relevance of carbonic anhydrase-IX in high-risk, early-stage cervical cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2010;116:452–458. doi: 10.1016/j.ygyno.2009.10.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Brockton NT, Klimowicz AC, Bose P, Petrillo SK, Konno M, Rudmik L, Dean M, Nakoneshny SC, Matthews TW, Chandarana S, Lau HY, Magliocco AM, Dort JC. High stromal carbonic anhydrase IX expression is associated with nodal metastasis and decreased survival in patients with surgically-treated oral cavity squamous cell carcinoma. Oral Oncol. 2012;48:615–622. doi: 10.1016/j.oraloncology.2012.01.018. [DOI] [PubMed] [Google Scholar]
  • 149.Atkins M, Regan M, McDermott D, Mier J, Stanbridge E, Youmans A, Febbo P, Upton M, Lechpammer M, Signoretti S. Carbonic anhydrase IX expression predicts outcome of interleukin 2 therapy for renal cancer. Clin Cancer Res. 2005;11:3714–3721. doi: 10.1158/1078-0432.CCR-04-2019. [DOI] [PubMed] [Google Scholar]
  • 150.Generali D, Berruti A, Brizzi MP, Campo L, Bonardi S, Wigfield S, Bersiga A, Allevi G, Milani M, Aguggini S, Gandolfi V, Dogliotti L, Bottini A, Harris AL, Fox SB. Hypoxia-inducible factor-1alpha expression predicts a poor response to primary chemoendocrine therapy and disease-free survival in primary human breast cancer. Clin Cancer Res. 2006;12:4562–4568. doi: 10.1158/1078-0432.CCR-05-2690. [DOI] [PubMed] [Google Scholar]
  • 151.Goonewardene TI, Sowter HM, Harris AL. Hypoxia-induced pathways in breast cancer. Microsc Res Tech. 2002;59:41–48. doi: 10.1002/jemt.10175. [DOI] [PubMed] [Google Scholar]
  • 152.Tafreshi NK, Bui MM, Bishop K, Lloyd MC, Enkemann SA, Lopez AS, Abrahams D, Carter BW, Vagner J, Grobmyer SR, Gillies RJ, Morse DL. Noninvasive detection of breast cancer lymph node metastasis using carbonic anhydrases IX and XII targeted imaging probes. Clin Cancer Res. 2012;18:207–219. doi: 10.1158/1078-0432.CCR-11-0238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zavada J, Zavadova Z, Zat’ovicova M, Hyrsl L, Kawaciuk I. Soluble form of carbonic anhydrase IX (CA IX) in the serum and urine of renal carcinoma patients. Br J Cancer. 2003;89:1067–1071. doi: 10.1038/sj.bjc.6601264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Li G, Feng G, Gentil-Perret A, Genin C, Tostain J. Serum carbonic anhydrase 9 level is associated with postoperative recurrence of conventional renal cell cancer. J Urol. 2008;180:510–513. doi: 10.1016/j.juro.2008.04.024. [DOI] [PubMed] [Google Scholar]
  • 155.Murakami Y, Kanda K, Tsuji M, Kanayama H, Kagawa S. MN/CA9 gene expression as a potential biomarker in renal cell carcinoma. BJU Int. 1999;83:743–747. doi: 10.1046/j.1464-410x.1999.00007.x. [DOI] [PubMed] [Google Scholar]
  • 156.Hyrsl L, Zavada J, Zavadova Z, Kawaciuk I, Vesely S, Skapa P. Soluble form of carbonic anhydrase IX (CAIX) in transitional cell carcinoma of urinary tract. Neoplasma. 2009;56:298–302. doi: 10.4149/neo_2009_04_29. [DOI] [PubMed] [Google Scholar]
  • 157.Kock L, Mahner S, Choschzick M, Eulenburg C, Milde-Langosch K, Schwarz J, Jaenicke F, Muller V, Woelber L. Serum carbonic anhydrase IX and its prognostic relevance in vulvar cancer. Int J Gynecol Cancer. 2011;21:141–148. doi: 10.1097/IGC.0b013e318204c34f. [DOI] [PubMed] [Google Scholar]
  • 158.Muller V, Riethdorf S, Rack B, Janni W, Fasching PA, Solomayer E, Aktas B, Kasimir-Bauer S, Zeitz J, Pantel K, Fehm T. Prospective evaluation of serum tissue inhibitor of metalloproteinase 1 and carbonic anhydrase IX in correlation to circulating tumor cells in patients with metastatic breast cancer. Breast Cancer Res. 2011;13:R71. doi: 10.1186/bcr2916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Woelber L, Mueller V, Eulenburg C, Schwarz J, Carney W, Jaenicke F, Milde-Langosch K, Mahner S. Serum carbonic anhydrase IX during first-line therapy of ovarian cancer. Gynecol Oncol. 2010;117:183–188. doi: 10.1016/j.ygyno.2009.11.029. [DOI] [PubMed] [Google Scholar]
  • 160.Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human cancer. Nat Rev Cancer. 2008;8:967–975. doi: 10.1038/nrc2540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Scheurer SB, Rybak JN, Rosli C, Neri D, Elia G. Modulation of gene expression by hypoxia in human umbilical cord vein endothelial cells: A transcriptomic and proteomic study. Proteomics. 2004;4:1737–1760. doi: 10.1002/pmic.200300689. [DOI] [PubMed] [Google Scholar]
  • 162.Steffens MG, Boerman OC, Oosterwijk-Wakka JC, Oosterhof GO, Witjes JA, Koenders EB, Oyen WJ, Buijs WC, Debruyne FM, Corstens FH, Oosterwijk E. Targeting of renal cell carcinoma with iodine-131-labeled chimeric monoclonal antibody G250. J Clin Oncol. 1997;15:1529–1537. doi: 10.1200/JCO.1997.15.4.1529. [DOI] [PubMed] [Google Scholar]
  • 163.Stillebroer AB, Oosterwijk E, Oyen WJ, Mulders PF, Boerman OC. Radiolabeled antibodies in renal cell carcinoma. Cancer Imaging. 2007;7:179–188. doi: 10.1102/1470-7330.2007.0025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Divgi CR, Pandit-Taskar N, Jungbluth AA, Reuter VE, Gonen M, Ruan S, Pierre C, Nagel A, Pryma DA, Humm J, Larson SM, Old LJ, Russo P. Preoperative characterisation of clear-cell renal carcinoma using iodine-124-labelled antibody chimeric G250 (124I-cG250) and PET in patients with renal masses: a phase I trial. Lancet Oncol. 2007;8:304–310. doi: 10.1016/S1470-2045(07)70044-X. [DOI] [PubMed] [Google Scholar]
  • 165.Reichert JM. Next generation and biosimilar monoclonal antibodies: essential considerations towards regulatory acceptance in Europe. February 3–4, 2011, Freiburg, Germany. MAbs. 2011;3:223–240. doi: 10.4161/mabs.3.3.15475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Li Y, Wang H, Oosterwijk E, Selman Y, Mira JC, Medrano T, Shiverick KT, Frost SC. Antibody-specific detection of CAIX in breast and prostate cancers. Biochem Biophys Res Commun. 2009;386:488–492. doi: 10.1016/j.bbrc.2009.06.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Chrastina A, Zavada J, Parkkila S, Kaluz S, Kaluzova M, Rajcani J, Pastorek J, Pastorekova S. Biodistribution and pharmacokinetics of 125I-labeled monoclonal antibody M75 specific for carbonic anhydrase IX, an intrinsic marker of hypoxia, in nude mice xenografted with human colorectal carcinoma. Int J Cancer. 2003;105:873–881. doi: 10.1002/ijc.11142. [DOI] [PubMed] [Google Scholar]
  • 168.Chrastina A, Pastorekova S, Pastorek J. Immunotargeting of human cervical carcinoma xenograft expressing CA IX tumor-associated antigen by 125I-labeled M75 monoclonal antibody. Neoplasma. 2003;50:13–21. [PubMed] [Google Scholar]
  • 169.Ahlskog JK, Schliemann C, Marlind J, Qureshi U, Ammar A, Pedley RB, Neri D. Human monoclonal antibodies targeting carbonic anhydrase IX for the molecular imaging of hypoxic regions in solid tumours. Br J Cancer. 2009;101:645–657. doi: 10.1038/sj.bjc.6605200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Vullo D, Franchi M, Gallori E, Pastorek J, Scozzafava A, Pastorekova S, Supuran CT. Carbonic anhydrase inhibitors. inhibition of cytosolic isozymes I and II and transmembrane, cancer-associated isozyme IX with anions. J Enzyme Inhib Med Chem. 2003;18:403–406. doi: 10.1080/1475636031000138732. [DOI] [PubMed] [Google Scholar]
  • 171.Innocenti A, Vullo D, Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors: interactions of phenols with the 12 catalytically active mammalian isoforms (CA I-XIV) Bioorg Med Chem Lett. 2008;18:1583–1587. doi: 10.1016/j.bmcl.2008.01.077. [DOI] [PubMed] [Google Scholar]
  • 172.Maresca A, Temperini C, Vu H, Pham NB, Poulsen SA, Scozzafava A, Quinn RJ, Supuran CT. Non-zinc mediated inhibition of carbonic anhydrases: coumarins are a new class of suicide inhibitors. J Am Chem Soc. 2009;131:3057–3062. doi: 10.1021/ja809683v. [DOI] [PubMed] [Google Scholar]
  • 173.Pastorekova S, Casini A, Scozzafava A, Vullo D, Pastorek J, Supuran CT. Carbonic anhydrase inhibitors: the first selective, membrane-impermeant inhibitors targeting the tumor-associated isozyme IX. Bioorg Med Chem Lett. 2004;14:869–873. doi: 10.1016/j.bmcl.2003.12.029. [DOI] [PubMed] [Google Scholar]
  • 174.Casey JR, Morgan PE, Vullo D, Scozzafava A, Mastrolorenzo A, Supuran CT. Carbonic anhydrase inhibitors. Design of selective, membrane-impermeant inhibitors targeting the human tumor-associated isozyme IX. J Med Chem. 2004;47:2337–2347. doi: 10.1021/jm031079w. [DOI] [PubMed] [Google Scholar]
  • 175.Winum JY, Pastorekova S, Jakubickova L, Montero JL, Scozzafava A, Pastorek J, Vullo D, Innocenti A, Supuran CT. Carbonic anhydrase inhibitors: synthesis and inhibition of cytosolic/tumor-associated carbonic anhydrase isozymes I, II, and IX with bis-sulfamates. Bioorg Med Chem Lett. 2005;15:579–584. doi: 10.1016/j.bmcl.2004.11.058. [DOI] [PubMed] [Google Scholar]
  • 176.Pacchiano F, Carta F, McDonald PC, Lou Y, Vullo D, Scozzafava A, Dedhar S, Supuran CT. Ureido-substituted benzenesulfonamides potently inhibit carbonic anhydrase IX and show antimetastatic activity in a model of breast cancer metastasis. J Med Chem. 2011;54:1896–1902. doi: 10.1021/jm101541x. [DOI] [PubMed] [Google Scholar]
  • 177.Dubois L, Lieuwes NG, Maresca A, Thiry A, Supuran CT, Scozzafava A, Wouters BG, Lambin P. Imaging of CA IX with fluorescent labelled sulfonamides distinguishes hypoxic and (re)-oxygenated cells in a xenograft tumour model. Radiother Oncol. 2009;92:423–428. doi: 10.1016/j.radonc.2009.06.019. [DOI] [PubMed] [Google Scholar]
  • 178.Cecchi A, Hulikova A, Pastorek J, Pastorekova S, Scozzafava A, Winum JY, Montero JL, Supuran CT. Carbonic anhydrase inhibitors. Design of fluorescent sulfonamides as probes of tumor-associated carbonic anhydrase IX that inhibit isozyme IX-mediated acidification of hypoxic tumors. J Med Chem. 2005;48:4834–4841. doi: 10.1021/jm0501073. [DOI] [PubMed] [Google Scholar]
  • 179.Dubois L, Douma K, Supuran CT, Chiu RK, van Zandvoort MA, Pastorekova S, Scozzafava A, Wouters BG, Lambin P. Imaging the hypoxia surrogate marker CA IX requires expression and catalytic activity for binding fluorescent sulfonamide inhibitors. Radiother Oncol. 2007;83:367–373. doi: 10.1016/j.radonc.2007.04.018. [DOI] [PubMed] [Google Scholar]
  • 180.Thiry A, Supuran CT, Masereel B, Dogne JM. Recent developments of carbonic anhydrase inhibitors as potential anticancer drugs. J Med Chem. 2008;51:3051–3056. doi: 10.1021/jm701526d. [DOI] [PubMed] [Google Scholar]
  • 181.Winum JY, Rami M, Scozzafava A, Montero JL, Supuran C. Carbonic anhydrase IX: a new druggable target for the design of antitumor agents. Med Res Rev. 2008;28:445–463. doi: 10.1002/med.20112. [DOI] [PubMed] [Google Scholar]
  • 182.Ahlskog JK, Dumelin CE, Trussel S, Marlind J, Neri D. In vivo targeting of tumor-associated carbonic anhydrases using acetazolamide derivatives. Bioorg Med Chem Lett. 2009;19:4851–4856. doi: 10.1016/j.bmcl.2009.06.022. [DOI] [PubMed] [Google Scholar]
  • 183.Helmlinger G, Sckell A, Dellian M, Forbes NS, Jain RK. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin Cancer Res. 2002;8:1284–1291. [PubMed] [Google Scholar]
  • 184.Groves K, Bao B, Zhang J, Handy E, Kennedy P, Cuneo G, Supuran CT, Yared W, Peterson JD, Rajopadhye M. Synthesis and evaluation of near-infrared fluorescent sulfonamide derivatives for imaging of hypoxia-induced carbonic anhydrase IX expression in tumors. Bioorg Med Chem Lett. 2012;22:653–657. doi: 10.1016/j.bmcl.2011.10.058. [DOI] [PubMed] [Google Scholar]
  • 185.Akurathi V, Dubois L, Lieuwes NG, Chitneni SK, Cleynhens BJ, Vullo D, Supuran CT, Verbruggen AM, Lambin P, Bormans GM. Synthesis and biological evaluation of a 99mTc-labelled sulfonamide conjugate for in vivo visualization of carbonic anhydrase IX expression in tumor hypoxia. Nucl Med Biol. 2010;37:557–564. doi: 10.1016/j.nucmedbio.2010.02.006. [DOI] [PubMed] [Google Scholar]
  • 186.Surfus JE, Hank JA, Oosterwijk E, Welt S, Lindstrom MJ, Albertini MR, Schiller JH, Sondel PM. Anti-renal-cell carcinoma chimeric antibody G250 facilitates antibody-dependent cellular cytotoxicity with in vitro and in vivo interleukin-2-activated effectors. J Immunother Emphasis Tumor Immunol. 1996;19:184–191. doi: 10.1097/00002371-199605000-00003. [DOI] [PubMed] [Google Scholar]
  • 187.Siebels M, Rohrmann K, Oberneder R, Stahler M, Haseke N, Beck J, Hofmann R, Kindler M, Kloepfer P, Stief C. A clinical phase I/II trial with the monoclonal antibody cG250 (RENCAREX(R)) and interferon-alpha-2a in metastatic renal cell carcinoma patients. World J Urol. 2011;29:121–126. doi: 10.1007/s00345-010-0570-2. [DOI] [PubMed] [Google Scholar]
  • 188.Davis ID, Wiseman GA, Lee FT, Gansen DN, Hopkins W, Papenfuss AT, Liu Z, Moynihan TJ, Croghan GA, Adjei AA, Hoffman EW, Ingle JN, Old LJ, Scott AM. A phase I multiple dose, dose escalation study of cG250 monoclonal antibody in patients with advanced renal cell carcinoma. Cancer Immun. 2007;7:13. [PMC free article] [PubMed] [Google Scholar]
  • 189.Zatovicova M, Jelenska L, Hulikova A, Csaderova L, Ditte Z, Ditte P, Goliasova T, Pastorek J, Pastorekova S. Carbonic anhydrase IX as an anticancer therapy target: preclinical evaluation of internalizing monoclonal antibody directed to catalytic domain. Curr Pharm Des. 2010;16:3255–3263. doi: 10.2174/138161210793429832. [DOI] [PubMed] [Google Scholar]
  • 190.Murri-Plesko MT, Hulikova A, Oosterwijk E, Scott AM, Zortea A, Harris AL, Ritter G, Old L, Bauer S, Swietach P, Renner C. Antibody inhibiting enzymatic activity of tumour-associated carbonic anhydrase isoform IX. Eur J Pharmacol. 2011;657:173–183. doi: 10.1016/j.ejphar.2011.01.063. [DOI] [PubMed] [Google Scholar]
  • 191.Brouwers AH, van Eerd JE, Frielink C, Oosterwijk E, Oyen WJ, Corstens FH, Boerman OC. Optimization of radioimmunotherapy of renal cell carcinoma: labeling of monoclonal antibody cG250 with 131I, 90Y, 177Lu, or 186Re. J Nucl Med. 2004;45:327–337. [PubMed] [Google Scholar]
  • 192.Stillebroer AB, Zegers CM, Boerman OC, Oosterwijk E, Mulders PF, O’Donoghue JA, Visser EP, Oyen WJ. Dosimetric analysis of 177Lu-cG250 radioimmunotherapy in renal cell carcinoma patients: correlation with myelotoxicity and pretherapeutic absorbed dose predictions based on 111In-cG250 imaging. J Nucl Med. 2012;53:82–89. doi: 10.2967/jnumed.111.094896. [DOI] [PubMed] [Google Scholar]
  • 193.Petrul HM, Schatz CA, Kopitz CC, Adnane L, McCabe TJ, Trail P, Ha S, Chang YS, Voznesensky A, Ranges G, Tamburini PP. Therapeutic mechanism and efficacy of the antibody-drug conjugate BAY 79-4620 targeting human carbonic anhydrase 9. Mol Cancer Ther. 2012;11:340–349. doi: 10.1158/1535-7163.MCT-11-0523. [DOI] [PubMed] [Google Scholar]
  • 194.Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol. 2006;24:e20–22. doi: 10.1200/JCO.2006.05.9964. [DOI] [PubMed] [Google Scholar]
  • 195.Dittrich C, Zandvliet AS, Gneist M, Huitema AD, King AA, Wanders J. A phase I and pharmacokinetic study of indisulam in combination with carboplatin. Br J Cancer. 2007;96:559–566. doi: 10.1038/sj.bjc.6603606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Buller F, Steiner M, Frey K, Mircsof D, Scheuermann J, Kalisch M, Buhlmann P, Supuran CT, Neri D. Selection of Carbonic Anhydrase IX Inhibitors from One Million DNA-Encoded Compounds. ACS Chem Biol. 2011;6:336–344. doi: 10.1021/cb1003477. [DOI] [PubMed] [Google Scholar]
  • 197.Stiti M, Cecchi A, Rami M, Abdaoui M, Barragan-Montero V, Scozzafava A, Guari Y, Winum JY, Supuran CT. Carbonic anhydrase inhibitor coated gold nanoparticles selectively inhibit the tumor-associated isoform IX over the cytosolic isozymes I and II. J Am Chem Soc. 2008;130:16130–16131. doi: 10.1021/ja805558k. [DOI] [PubMed] [Google Scholar]
  • 198.Tureci O, Sahin U, Vollmar E, Siemer S, Gottert E, Seitz G, Parkkila AK, Shah GN, Grubb JH, Pfreundschuh M, Sly WS. Human carbonic anhydrase XII: cDNA cloning, expression, and chromosomal localization of a carbonic anhydrase gene that is overexpressed in some renal cell cancers. Proc Natl Acad Sci U S A. 1998;95:7608–7613. doi: 10.1073/pnas.95.13.7608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Kivela A, Parkkila S, Saarnio J, Karttunen TJ, Kivela J, Parkkila AK, Waheed A, Sly WS, Grubb JH, Shah G, Tureci O, Rajaniemi H. Expression of a novel transmembrane carbonic anhydrase isozyme XII in normal human gut and colorectal tumors. Am J Pathol. 2000;156:577–584. doi: 10.1016/S0002-9440(10)64762-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Parkkila S, Parkkila AK, Saarnio J, Kivela J, Karttunen TJ, Kaunisto K, Waheed A, Sly WS, Tureci O, Virtanen I, Rajaniemi H. Expression of the membrane-associated carbonic anhydrase isozyme XII in the human kidney and renal tumors. J Histochem Cytochem. 2000;48:1601–1608. doi: 10.1177/002215540004801203. [DOI] [PubMed] [Google Scholar]
  • 201.Kivela AJ, Parkkila S, Saarnio J, Karttunen TJ, Kivela J, Parkkila AK, Pastorekova S, Pastorek J, Waheed A, Sly WS, Rajaniemi H. Expression of transmembrane carbonic anhydrase isoenzymes IX and XII in normal human pancreas and pancreatic tumours. Histochem Cell Biol. 2000;114:197–204. doi: 10.1007/s004180000181. [DOI] [PubMed] [Google Scholar]
  • 202.Kivela AJ, Parkkila S, Saarnio J, Karttunen TJ, Kivela J, Parkkila AK, Bartosova M, Mucha V, Novak M, Waheed A, Sly WS, Rajaniemi H, Pastorekova S, Pastorek J. Expression of von Hippel-Lindau tumor suppressor and tumor-associated carbonic anhydrases IX and XII in normal and neoplastic colorectal mucosa. World J Gastroenterol. 2005;11:2616–2625. doi: 10.3748/wjg.v11.i17.2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Brennan DJ, Jirstrom K, Kronblad A, Millikan RC, Landberg G, Duffy MJ, Ryden L, Gallagher WM, O’Brien SL. CA IX is an independent prognostic marker in premenopausal breast cancer patients with one to three positive lymph nodes and a putative marker of radiation resistance. Clin Cancer Res. 2006;12:6421–6431. doi: 10.1158/1078-0432.CCR-06-0480. [DOI] [PubMed] [Google Scholar]
  • 204.Span PN, Bussink J, Manders P, Beex LV, Sweep CG. Carbonic anhydrase-9 expression levels and prognosis in human breast cancer: association with treatment outcome. Br J Cancer. 2003;89:271–276. doi: 10.1038/sj.bjc.6601122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Watson PH, Chia SK, Wykoff CC, Han C, Leek RD, Sly WS, Gatter KC, Ratcliffe P, Harris AL. Carbonic anhydrase XII is a marker of good prognosis in invasive breast carcinoma. Br J Cancer. 2003;88:1065–1070. doi: 10.1038/sj.bjc.6600796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Marr A, Markert A, Altmann A, Askoxylakis V, Haberkorn U. Biotechnology techniques for the development of new tumor specific peptides. Methods. 2011;55:215–222. doi: 10.1016/j.ymeth.2011.05.002. [DOI] [PubMed] [Google Scholar]
  • 207.Askoxylakis V, Garcia-Boy R, Rana S, Kramer S, Hebling U, Mier W, Altmann A, Markert A, Debus J, Haberkorn U. A new peptide ligand for targeting human carbonic anhydrase IX, identified through the phage display technology. PLoS One. 2010;5:e15962. doi: 10.1371/journal.pone.0015962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Rana S, Nissen F, Marr A, Markert A, Altmann A, Mier W, Debus J, Haberkorn U, Askoxylakis V. Optimization of a novel peptide ligand targeting human carbonic anhydrase IX. PLoS One. 2012;7:e38279. doi: 10.1371/journal.pone.0038279. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES