Next Article in Journal
Ring-like (Donut-Shaped) Intracranial Aneurysms: A Warning Morphology of Mural Jet Flow and Pre-Rupture Instability
Previous Article in Journal
The Impact of Axial CT Level Selection on Grading Trochlear Dysplasia Using Dejour Classification
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Tumor pH in Breast Cancer Imaging: Biology, Diagnostic Applications, and Emerging Techniques

1
Department of Radiology, Jacobi Medical Center, Bronx, NY 10461, USA
2
Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
*
Author to whom correspondence should be addressed.
Diagnostics 2026, 16(1), 76; https://doi.org/10.3390/diagnostics16010076 (registering DOI)
Submission received: 1 November 2025 / Revised: 15 December 2025 / Accepted: 17 December 2025 / Published: 25 December 2025
(This article belongs to the Special Issue Advances in Breast Diagnostics)

Abstract

Breast cancer screening, while vital for reducing mortality, faces significant limitations in sensitivity and specificity, particularly in dense breasts. Current modalities primarily detect anatomical changes, often missing biologically aggressive tumors at their earliest stages. The altered metabolism of cancer cells establishes a characteristic inverted pH gradient that drives tumor invasion, metastasis, and treatment resistance. This makes tumor acidity a compelling, functional biomarker for early detection. This review synthesizes the emerging role of pH as a diagnostic biomarker and provides a critical evaluation of advanced imaging techniques for its non-invasive or minimal measurement. We detail the biological underpinnings of tumor acidosis, emphasizing its regulation through glycolytic reprogramming and dysregulated proton transport. Our analysis encompasses a broad spectrum of pH-sensitive imaging modalities, including magnetic resonance methods such as Chemical Exchange Saturation Transfer (CEST) MRI for extracellular pH mapping and multi-nuclear Magnetic Resonance Spectroscopy (MRS) using 1H, 31P, and 19F nuclei to probe various cellular compartments. Furthermore, we examine hyperpolarized 13C MRI for real-time metabolic flux imaging, where metrics such as the lactate-to-pyruvate ratio demonstrate significant predictive value for treatment response. The review also assesses optical and photoacoustic imaging techniques, which offer high sensitivity but are often constrained to superficial tumors. Imaging tumor pH provides a powerful functional window into the earliest metabolic shifts in breast cancer, far preceding macroscopic anatomical changes. The ongoing development and evidence support the role of the pH-sensitive imaging techniques in diagnosis, lesion characterization, and therapy. Additionally, it holds promise for supplementing breast cancer screening by enabling earlier, more specific detection and personalized risk stratification, ultimately aiming to improve patient outcomes.

1. Introduction

Despite widespread adoption of breast cancer screening, tumors can still remain underdetected, contributing to continued morbidity and mortality [1]. The most common cancer among women worldwide is breast cancer, with 2.3 million new cases annually accounting for 30% of cancers in women with rising incidence rates [2,3]. To reduce mortality by early detection of cancer, the US Preventive Services Task Force (USPSTF) recommends biennial screening with digital mammography (DM) and digital breast tomosynthesis (DBT, also known as 3D-mammography) for women aged 40 to 74. Supplemental modalities such as breast ultrasonography, magnetic resonance imaging (MRI) and contrast enhanced mammography are advised for women with high lifetime risk based on familial and personal factors [4]. While patients with dense breasts are challenging to screen with DM and DBT, and there is ongoing research into the use of supplemental screening techniques such as MRI in these patients, per the USPSTF there is currently insufficient evidence to recommend this modality for screnning. For diagnosis, additional modalities such as dynamic contrast enhanced MRI (DCE-MRI) which evaluates the angiogenic changes in malignancy, have shown increased specificity for the detection of breast cancer compared to non-malignant processes [5]. DCE-MRI can also be used to monitor treatment response in patients undergoing neoadjuvant chemotherapy, though differentiating residual disease from treatment changes remains challenging [6].
Not all women with high mammographic breast density (MBD) will develop breast cancer, and some women with low MBD will, which is why another marker for breast cancer risk is needed [7,8]. All of the aforementioned imaging modalities rely on identification of morphological changes in cancer, and as a result they have notable limitations: reduced sensitivity and specificity in dense breasts, risk of false positives and false negatives, and potential for overdiagnosis, all of which can lead to increased testing , invasive follow-up procedures, and psychological distress [4,9,10,11,12]. Moreover, by the time morphological abnormalities are visible, some tumors may already exhibit aggressive behaviour [13]. This underscores the need for screening approaches capable of detecting cancer earlier and with greater biological specificity, particularly in patients with dense breast tissue. One promising avenue is functional and metabolic imaging, such as targeting tumor pH. Cancer cells, unlike normal cells, exhibit altered metabolism to support intensive growth and turnover characterized by a more basic intracellular pH and more acidic extracellular pH due to the Warburg effect [14,15,16,17,18]. The Warburg effect is a well-known phenomenon in which tumors, even in the presence of sufficient oxygen, produce lactate through the glycolytic pathways rather than oxidative phosphorylation [18]. An acidic pH in breast cancer cells has been seen to contribute to increased invasive growth, migration, aggressiveness, metastasis, and poor prognosis, making pH a compelling biomarker for early breast cancer screening and detection [19,20,21,22,23,24,25].
Recent improvements in advanced imaging modalities can help improve our ability to detect breast cancer earlier by overcoming the limitations of traditional techniques, and may provide evidence for future improvements in screening and diagnostic guidelines, particularly for patients with dense breast tissue. This review synthesizes current evidence and emerging advances in pH-sensitive breast imaging, comparing these innovations with established modalities to evaluate their feasibility for earlier cancer detection.

2. Tumor pH Biology and Its Role in Breast Cancer

2.1. Why pH Is a Hallmark of Cancer?

Tumor pH is a defining metabolic hallmark of tumors, characterized by an inverted pH gradient due to an altered metabolism, a fundamental reversal from physiology that Koltai T termed “the cancer pH paradigm” [26,27,28,29,30]. Typically, extracellular pH (pHe) ranges from 7.3 and 7.4 while intracellular pH (pHi) ranges from 7.0 to 7.3 [31,32]. The reversal of this pHi/pHe gradient in cancer is a result of interplay between glycolytic reprogramming and dysregulated proton dynamics [18,33,34]. The Warburg effect, describing tumor cells’ increased oxidative phosphorylation, drives lactate and proton (H+) overproduction; these acids are extruded via a Na+/H+ exchanger (NHE), the H+-lactate co-transporter, monocarboxylate transporters (MCTs), and the H+-ATPase (H+ pump), which lead to the secretion of H+ creating an acidic tumor microenvironment and an alkaline cytosolic environment [33,34]. The change in pHi promotes cell proliferation and cell survival by limiting apoptosis, which is associated with intracellular acidification [35,36,37,38,39,40]. Extracellular acidosis actively drives malignancy by promoting invasion (via MMP-9 activation), evasion of immune surveillance, and enhancing chemoresistance, among other effects [28,41,42,43,44,45,46,47,48]. In breast cancer, this pH inversion critically influences tumor aggressiveness, metastatic potential, and treatment response, while also offering opportunities for early detection strategies. Detecting tumor acidity through pH-sensitive imaging could identify cancers at their earliest metabolic stages, long before structural changes appear on traditional mammograms or DBT.

2.2. Acidic Shift in Breast Cancer

In cancer, there are multiple pathways that contribute to the regulation of pHe/pHi [49,50]. Cancer cells tend to have a higher alkaline pHi (>7.4) and lower acidic pHe (6.7–7.1) compared to normal cells [15]. This shift is not a passive occurrence but an active, orchestrated process driven by metabolic reprogramming and precise molecular regulation. In breast cancer cells, the acidity of the extracellular environment is attributed to multiple causes. The Warburg effect (aerobic glycolysis) is initiated by oncogenic signaling and a failure of energy-sensing checkpoints, particularly the loss of the LKB1-AMPK pathway, which allows cells with inefficient, acid-producing metabolism to evade growth arrest and continue proliferating [51,52]. Malignant tumors have increased tumor pHi than lower grade tumors [53]. The massive production of lactic acid and protons from glycolysis creates a critical need for acid extrusion. This is achieved through the coordinated upregulation of specific transporters on the plasma membrane or through phagosomes [54,55]. Monocarboxylate transporters MCT1 and MCT4 (SLC16A1, SLC16A3) are pivotal, co-exporting lactate alongside protons (H+) into the extracellular space, directly acidifying the tumor microenvironment [56,57,58]. Their expression and membrane localization are dependent on their association with the chaperone protein CD147, which is itself linked to poor prognosis [59,60,61]. Simultaneously, proton pumps, specifically the plasma membrane H+-ATPase, operate aggressively and uniformly across the cell surface to pump protons out, further contributing to the severe extracellular acidification [54,62]. Additionally, phagosomes can also contribute as they lead to increased leakage of protons [55,63,64].
To survive the acidic environment they are creating and to maintain the alkaline pHi necessary for cell proliferation, cancer cells concurrently activate acid-neutralizing import mechanisms. The Na+/H+ exchanger (NHE1) extrudes intracellular H+ in exchange for extracellular Na+, while the Na+, HCO3 cotransporter NBCn1 (SCL4A7) imports bicarbonate ions, which buffer the cytosol [65,66,67]. Lowered cancer cell proliferation and reduced breast tumor growth is seen when the expression of NBCn1 is disrupted, but elevated levels are associated with shorter survival and increased metastasis [68,69]. In contrast, NHE1 is linked to promoting motility in ErbB2-positive breast cancer cells, stimulating the epithelial-to-mesenchymal transition (EMT) and contributing to chemotherapy resistance [65,66,70,71,72]. Additionally, a loss of the extracellular HCO3 sensing protein receptor protein tyrosine phosphatase RPTPγ is observed in premalignant lesions of the breast. The upregulation or downregulation of these transporters is not random; it is directly driven by oncogenic pathways. For example, the tyrosine kinase ErbB2 (HER2), a key driver in certain breast cancer subtypes, enhances the expression and activity of both NHE1 and NBCn1 via the ERK-RSK signaling pathway. This direct link ensures that the pH regulatory machinery is integrated with the core proliferative signals of the cancer cell [73,74,75].
The resulting inverted pH gradient is profoundly functional, promoting invasion and metastasis. Intracellular alkalinization protects the cancer cell from acid-induced damage and promotes proliferation [54,62]. Conversely, extracellular acidification remodels the tumor microenvironment by activating secreted enzymes, such as cathepsins, which degrade the extracellular matrix (ECM), thereby clearing a path for invasion. Furthermore, the acidic pHe impairs immune cell function, creating an immunosuppressive shield [76]. Thus, the coordinated activity of MCTs, H+-ATPases, NHE1, and NBCn1, all of which are regulated by oncogenic drivers, creates a permissive environment for tumor growth, invasion, and ultimately, poor patient outcomes (Figure 1).

3. Imaging Techniques for pH Detection in Breast Cancer

Due to changes in pH within the tumor, various imaging techniques are being evaluated to measure in vivo acidosis, including magnetic resonance-based methods (1H-MRS, 31P-MRS, 19F-MRS, and hyperpolarized 13C MRI), CEST imaging, optical/fluorescence imaging, photoacoustic imaging (PAI), and Fludeoxyglucose-18 positron emission tomography scan (FDG-PET). In this section, we review the imaging techniques listed above, in the order presented, and their application in breast cancer.

3.1. MR Spectroscopy Methods

Magnetic resonance spectroscopy (MRS) techniques offer the ability to derive metabolic information without the use of ionizing radiation [77,78]. Unlike standard MRI, which relies on the bulk signal of all protons, MRS techniques rely on the fact that different proton groups in molecules will resonate at slightly different frequencies in a magnetic field [79]. MRS techniques seek to identify and quantify different cellular chemicals based on the relative signals detected at these resonance offsets, providing quantitative insight into tissue metabolism in vivo [78,80]. MRS operates on the same scanner as an MRI but is uniquely designed to detect and quantify the concentrations of specific endogenous metabolites within living tissue [79,80]. By revealing these metabolic alterations, MRS can identify pathological processes even before any structural damage becomes visible [78]. However, the technique presents significant technical challenges, as it is highly sensitive to magnetic field imperfections, must detect molecules present in very low concentrations, and requires complex analysis to interpret the resulting spectral data [77,78].
a.
Proton Magnetic Resonance Spectroscopy (1H-MRS)
1H-MRS represents a valuable approach for assessing tumor microenvironment parameters through detection of both endogenous metabolites and exogenous probes, leveraging the high natural abundance (99.98%) and superior sensitivity of the proton nucleus in human tissue [78,81]. This technique has demonstrated clinical utility in breast cancer characterization through detection of elevated choline compounds (tCho), which serve as a biomarker of increased membrane turnover in malignant tissues, with diagnostic sensitivity and specificity ranging from 71 to 80% [82,83,84]. More recently, attention has turned to lactate as a hypoxia-associated metabolite, with the study demonstrating that lactate can be detected in the human breast noninvasively using 1H-MRS [85].
However, conventional 1H-MRS faces challenges in breast tissue due to the high concentration of lipids in breast tissue. Lipid resonances can overlap with many other relevant chemical compounds, including choline. Techniques used elsewhere in the body, such as placement of saturation bands or masking of lipid-containing tissues, can be difficult to implement in the breast [86,87]. Double-quantum filtered (DQF) MRS methods have been developed to suppress these interfering lipid signals, enabling the specific detection of lactate and revealing significantly higher levels in grade III compared to grade II breast lesions, which supports its potential as a biomarker of tumor aggression [87,88,89,90,91,92,93].
For direct measurement of extracellular pH (pHe), exogenous imidazole-based probes such as (±)2-imidazole-1-yl-3-ethoxycarbonylpropionic acid (IEPA) and ISUCA have been developed, featuring pH-sensitive 1H resonances in the 7–9 ppm range, a spectral region relatively free from endogenous interferences [94,95,96,97,98,99]. These compounds enable ratiometric measurements through pH-dependent chemical shifts of their H-2 proton relative to internal reference protons, allowing concentration-independent pH assessment. Preclinical studies in orthotopic breast cancer models have successfully generated pHe maps using these approaches, revealing acidic regions within tumors that correlate with aggressive features [95,100]. While 31P MRS offers a more direct way to monitor phospholipid metabolites, energy metabolites, and intracellular pH levels, the increased spectral resolution at higher field strengths (7 T versus 1.5 T) has enabled distinct detection of phosphoethanolamine (PE) metabolites from phosphocholine (PC) metabolites in patient studies [101,102,103,104,105]. Clinical observations show altered metabolite ratios in breast cancer patients, with elevated PE resonances compared to PC resonances, which may partially explain the tCho levels detected. Furthermore, alterations in pH levels have been observed clinically through split Pi resonances, with calculations yielding pH values of 7.5 and 6.9 that align with literature reports [106,107].
However, the technique faces challenges, including relatively small pH-dependent chemical shifts (approximately 0.7 ppm over the physiological range) that require high magnetic field strengths (e.g., 3 T) for adequate resolution, and spatial resolution on the order of multiple millimeters that may be insufficient for characterizing heterogeneous tumor microenvironments [100]. Additionally, some imidazole-based probes exhibit buffering capacity that could potentially alter the pHe they are intended to measure, although recent developments in diimidazole compounds aim to address these limitations with enhanced sensitivity and reduced buffering capacity [94,108]. Despite these challenges, the technique’s compatibility with standard clinical MRI systems and its ability to simultaneously assess multiple metabolic parameters position it as a promising multiparametric approach for evaluating breast cancer.
b.
Phosphorus MRS (31P-MRS)
Phosphate compounds play a central role in cellular energy metabolism; all energetic processes in human tissue cells involve phosphate-containing metabolites such as phosphocreatine (PCr), inorganic phosphate (Pi), and adenosine triphosphate (α-β-γ-ATP) [109]. Phosphorus MR Spectroscopy (31P-MRS) relies on the same phenomenon of difference in resonance frequencies of phosphorus when it is bound in different chemicals. 31P-MRS can measure intracellular pH by assessing the chemical shift difference between different phosphorus chemical compounds, with this difference changing in response to pH; this technique has been used to assess pH changes in multiple tissues and tumors [110]. Inorganic Pi is most affected by changes in pH, particularly in tumors, whereas PCr is used as a reference since it has a relatively stable resonance position across physiologic pH ranges [111,112,113] (Figure 2). Studies on 3 T MRI have shown an improved signal-to-noise ratio and spectral resolution in the human breast in vivo. Most studies have focused on the quantification of phosphorus metabolites in breast cancers as compared to normal tissue, with a focus on changes of these metabolites under chemotherapy, with an increase in pHi following treatment [114,115,116,117,118,119]. While MRS can be used to study the intracellular pH changes, which are the initial changes that occur in cancer cells, further studies have been conducted to assess the extracellular pH [120]. 31P MRS of 3-aminopropylphosphate (3-APP), a non-toxic, membrane-impermeant compound, is used to measure the extracellular pH of the tumor. It has been demonstrated to show changes in breast cancer cell lines [27]. A major benefit of 31P-MRS is the lack of need for water suppression to detect phosphorus metabolites, unlike in vivo metabolite detection with 1H-MRS [119]. Limitations of a 31P-MRS include lower sensitivity as Pi concentrations are generally lower than proton concentrations, and thus it is essential to use either slower or lower resolution techniques to acquire sufficiently high SNR for pHe quantification [110].
c.
Fluoride MRS (19F-MRS)
Fluoride MR spectroscopy (19F-MRS) has emerged as a powerful technique for probing the tumor microenvironment, particularly extracellular pH, due to the near absence of endogenous fluorine background in biological tissues [121,122,123]. This lack of background signal allows for unambiguous detection of exogenous fluorinated probes, while the nucleus’s high gyromagnetic ratio and large chemical shift dispersion (~300 ppm) provide excellent spectral resolution [121]. The development of effective 19F-MRS pH indicators requires careful optimization of pharmacokinetic properties, including a pKa within the physiological range, high sensitivity and specificity, low toxicity, efficient cellular delivery, and appropriate membrane permeability characteristics [120,122,123,124,125].
Several classes of fluorinated probes have been developed for pH sensing. Early approaches utilized fluorinated alanines, which demonstrated pH-dependent chemical shifts but were limited to perfused cell systems and were not applied in vivo [126,127,128,129]. The fluoroaniline sulfonamide ZK-150471 has shown particular promise, demonstrating a superior signal-to-noise ratio and pH sensitivity compared to 31P-MRS probes, such as 3-APP [120,130].
Recent innovations focus on “smart” contrast agents and nanoscale delivery systems [131,132,133]. PEGylated nanogels containing perfluorocarbons exhibit pH-dependent size changes, enabling indirect pH estimation through 19F signal variations [134,135,136,137,138,139]. Molecular switches acting as ratiometric probes eliminate the need for external references, while paramagnetic relaxation enhancement strategies using Mn2+ ions have improved signal intensity by over five-fold [135,137,140,141]. Liposil-encapsulated fluorinated ionic liquids represent another advancement, enabling multiplexed 19F MRI with enhanced relaxation properties [141].
Despite these advantages, 19F MRS faces challenges including probe instability, nonspecific accumulation due to hydrophobicity, and limited spatial resolution despite high field strengths [130,134]. While extracellular pH measurement is well-established, simultaneous assessment of intracellular pH remains challenging. Nevertheless, the continuous development of novel fluorinated compounds and encapsulation strategies positions 19F MRS as a valuable complementary approach to proton-based methods for characterizing tumor acidosis in breast cancer research and potential clinical applications [134,137].

3.2. Hyperpolarized Carbon-13 (13C-MRI)

Hyperpolarized carbon-13 (13C) magnetic resonance imaging (MRI) represents an advanced metabolic imaging technique that enables real-time, non-invasive assessment of tissue pH. The method enhances the signal of 13C-labeled compounds by cooling a 13C-enriched sample with a polarizing agent to near absolute zero in the presence of strong magnetic fields (3.35—7 T) for 2 h, and then transferring polarization via microwave irradiation to improve visualization of metabolic activity. However, this technique requires specialized equipment for polarization and rapid quality control, necessitating dedicated multinuclear MRI scanners with custom 13C coils. These technical and economic constraints have currently limited its widespread clinical implementation [142].
Two primary approaches facilitate pH quantification using hyperpolarized 13C MRI. The first method utilizes 13C-bicarbonate, which undergoes pH-dependent equilibrium with 13CO2 via carbonic anhydrase catalysis, enabling quantitative mapping of extracellular pH through signal intensity ratios. The second approach employs [1-13C] pyruvate, whose metabolic conversion to 13CO2 in highly aerobic tissues permits similar ratio-based calculations primarily reflecting intracellular pH. Since both compounds are endogenous metabolites, these techniques show significant potential for clinical translation in mapping pH distributions in human cancers [143].
In breast cancer applications, the hyperpolarized 13C-lactate signal has demonstrated utility as a potential biomarker for tumor grading, with detectable signals in triple-negative and high-grade tumors but absence in lower-grade malignancies [144,145]. The lactate-to-pyruvate ratio (Lac/Pyr) correlates strongly with HIF1α expression, linking metabolic activity to hypoxia and tumor volume, while also showing associations with lactate dehydrogenase A (LDHA) and monocarboxylate transporter 1 (SLC16A1/MCT1) expressions [146].
The technology shows particular promise for assessing treatment response, detecting metabolic alterations within 7–11 days post-treatment. Increases in Lac/Pyr ratio ≥ 20% following therapy have demonstrated predictive value for pathological complete response (pCR) [146,147]. However, data interpretation requires careful consideration, as the 13C-lactate signal reflects complex interactions involving substrate delivery, transporter expression, and enzymatic activity, rather than just lactate production. This complexity is evidenced by paradoxical observations such as increased lactate conversion following pazopanib treatment [148].
Recent technological innovations address limitations of conventional polarization methods. Novel approaches, such as Spin-Lock Induced Crossing with Signal Amplification by Reversible Exchange (SLIC-SABRE), enable the rapid generation of hyperpolarized [1-13C] pyruvate within minutes at a reduced cost, facilitating high-throughput studies. Applications in transgenic breast cancer models have successfully identified elevated lactate metabolism and metabolic subcompartments corresponding to histological profiles, demonstrating potential for overcoming current implementation barriers [149].
While challenges remain regarding technical complexity, cost, and data interpretation, ongoing advancements in hyperpolarized 13C MRI position it as a transformative approach for non-invasive pH monitoring in breast cancer management. The capability to track spatial and temporal pH dynamics provides unique insights into tumor heterogeneity and treatment response, offering significant potential for advancing personalized cancer care [144].

3.3. Chemical Exchange Saturation Transfer (CEST) MRI

Chemical exchange saturation transfer (CEST) imaging refers to a method for measuring low-concentration solutes through the phenomenon of magnetization transfer (MT). In MT, a specific proton group (e.g., amides) is stimulated at its resonance frequency using radiofrequency (RF) pulses, saturating its magnetization. This magnetization is transferred to the surrounding water due to the exchange of the excited protons with protons in bulk water. Repeated cycles of stimulation and transfer will lead to a build-up of signal in bulk water, which cannot be saturated, given its concentration is orders of magnitude higher than the stimulated proton groups. This increased signal can then be more easily detected [150,151,152,153,154]. The chemical exchange of saturated protons with protons of water is pH-dependent, and therefore, measurement of the rate of transfer can be used to estimate tissue pH [155] (Figure 3). CEST imaging has proven effective in detecting acidic pH of breast tumors and revealing their metastatic potential, as the CEST effect diminishes at higher, more basic pH levels due to an excessively rapid exchange rate. CEST is broadly categorized into two types: endogenous and exogenous CEST [156,157,158,159].
a.
Endogenous CEST:
This approach focuses on the stimulation of chemical groups endogenously found in tissues. The most established method is amide proton Transfer(APT) imaging, which focuses on amide protons (proteins and peptides) [159]. The exchange rate between the amide protons and water decreases with decreasing pH [155,160,161,162]. Since cancer cells exhibit higher intracellular protein concentrations due to rapid turnover, the APT signal is often increased [163,164,165]. Clinically, APT has demonstrated high diagnostic performance, distinguishing between benign and malignant breast lesions with an AUC as high as 0.959 [166,167]. Its signal shows a positive correlation with tumor stage and Ki-67 proliferation index, and decreases following effective neoadjuvant therapy [168]. A current limitation is that it has mostly been studied for characterizing larger lesions compared to smaller ones.
Other endogenous CEST methods target different protons, including hydroxyl groups (e.g., glycCEST and gagCEST, used in humans to distinguish tumors from surrounding tissue) and the amine groups of glutamate (gluCEST) [108,169,170]. While GluCEST has historically been used in the brain, recent work has shown promise in mouse models for monitoring early response to glutaminase inhibitors in triple-negative breast cancer [171]. Its clinical translation for breast cancers has been somewhat limited as it relies on high magnetic field strengths such as 7 T, which are not typically used in breast imaging [172].
b.
Exogenous CEST:
This method utilizes administered CEST contrast agents to specifically map the extracellular pH (pHe) of the tumor microenvironment [173]. It employs FDA-approved, non-ionic iodinated X-ray contrast agents known for high water solubility, low toxicity, and pH dependence, such as Iopromide, Ioversol [174,175,176]. Recent work in mouse models using iopamidol CEST found a correlation between breast tumor acidity and metastasis potential, and the relation with cancer with varying metabolic activity and HIF-1A expression [156,157,177]. A key advance is the development of acidoCEST MRI, a technique that leverages the differences in CEST effects of its two amide groups at varying pH levels. A ratio of the magnitude of these two CEST effects was found to be highly linearly correlated with pH in breast cancer [176]. This enables a ratiometric analysis method that calculates pHe independently of the agent’s concentration, a significant advantage for use in human subjects [174,178,179].
However, exogenous CEST faces challenges. Many pioneering studies have been conducted at 7 T, which is not widely available even at academic institutions, and validation on clinical 3 T systems is therefore needed. The accurate measurement range is typically limited to pH < 7.2, and while many tumor microenvironments are at pH < 7.4, the technique remains sensitive to confounding variables like temperature, pH, contrast agent, and magnetic field (B0/B1) inhomogeneity, necessitating careful experimental control [180,181].

3.4. Optical and Fluorescent Probes for Tumor pH Detection

Optical imaging represents a powerful approach for detecting the acidic tumor microenvironment, leveraging light-based technologies to provide accurate, real-time data on biochemical properties, including extracellular pH (pHe) [182,183]. These methods are particularly valuable for their high sensitivity and versatility, though their application is primarily limited to superficial or accessible tumors [106,184,185,186,187]. A key advantage of these approaches is their capacity for ratiometric measurements, where the ratio of signals at different emission or excitation wavelengths enables pH quantification independent of probe concentration, photobleaching, or tissue depth variations. This capability for concentration-independent measurement significantly enhances accuracy and reliability in pH detection [188].
For in vivo applications, the near-infrared (NIR) range (650–900 nm) is particularly valuable due to reduced light absorption and scattering by biological tissues, allowing penetration to deeper depths of the tissue of up to 1 cm. Several optical techniques have been developed for pH sensing, each with distinct characteristics [108,176]. Optical sensors such as Optical coherence tomography (OCT) and photoacoustic imaging (PAI) are a non-invasive technique for detecting tumors [189].
OCT employs non-invasive light-based imaging to generate high-resolution, micrometer-scale images of tissue architecture. OCT takes advantage of the high spatial resolution of near-infrared light to detect micrometer-level reflectances in tissue, an optical analogue of ultrasound [190]. OCT has demonstrated particular utility in identifying early-stage breast malignancies that evade detection by conventional imaging modalities. Clinical investigations have validated OCT’s diagnostic capability, reporting sensitivity of 93% and specificity of 85% in distinguishing cancerous breast tissue, establishing its potential as an effective tool for early cancer detection [191]. Fluorescence imaging utilizes pH-sensitive dyes that alter their emission properties in response to changes in acidity. Optical coherence tomography (OCT), although not a direct pH sensor, provides high-resolution structural imaging that complements pH measurements. The development of various probes has been crucial for advancing optical pH detection, ranging from visible-light probes like seminaphthorhodafluor-1 (SNARF-1) for preclinical window chamber models to NIR fluorescent probes such as modified rhodamine bound to dextran (Dex-Me-pEPPR) and activatable nanoparticles that turn on fluorescence in acidic conditions [192,193]. The pH can be measured independently of concentration by assessing the ratio of fluorescence signals at different emission wavelengths or at different fluorescence lifetimes [21,184,185,186,192].
PAI has emerged as a particularly promising modality for deeper tissue pH sensing, utilizing probes designed with pH-dependent and pH-independent absorption peaks [182]. To overcome the depth limitations of optical methods like photoacoustic imaging (PAI) has emerged as a modality for deeper tissue pH sensing. PAI is a hybrid modality that combines optical imaging with ultrasound detection, providing high spatial and temporal resolution for imaging structures several centimeters deep [194,195]. This technique can identify tumors as small as 1–2 mm with a sensitivity of 91%. Systems including albumin-based nanoparticles with benzo-α-phenoxazine and IR825, or polymer nanoparticles containing SNARF-5F, enable pH quantification through ratioing of optoacoustic signals at different excitation wavelengths [196]. Despite their considerable promise, the clinical translation of optical and photoacoustic pH-sensing for broad breast cancer screening faces hurdles. The primary limitation remains depth penetration, which restricts these methods to superficial or accessible tumors [106,196]. Furthermore, challenges related to biocompatibility and probe stability, along with addressing the complexities of absolute pH quantification due to nonlinear response curves, must be addressed [108]. Consequently, these technologies are currently best suited for specialized applications such as intraoperative guidance or endoscopic procedures, where their high sensitivity and capacity for spatial pH mapping can be fully leveraged [108,194,195,196,197].

3.5. FDG-PET

18F-fluorodeoxyglucose (FDG) PET/CT is widely used in oncology for diagnosis, staging, and surveillance [198]. As a non-invasive measure of glycolytic activity, FDG PET reflects the Warburg effect and enhanced reliance on aerobic glycolysis, which is observed in many cancers [199,200]. In breast cancer, higher FDG uptake correlates with increased concentrations of glycolytic metabolites, including lactate (p = 0.001) and L-acetylcarnitine (p = 2 × 10−4), which contribute to the acidic tumor microenvironment [200]. Because tumor acidosis arises largely from elevated glycolysis, regions of high FDG uptake correspond to areas of increased acidity, making FDG-PET a practical surrogate for visualizing tumor pH and assessing metabolic aggressiveness.
To more directly image tumor acidity, pH-low insertion peptides (pHLIPs) labeled with 89Zr have been developed as PET tracers that target acidic environments independently of molecular surface markers [201]. 89-Zr-DFO-Cys-Var3 is a PET probe that demonstrates uniform tumor staining and highlights acidified zones at the tumor–stroma interface. pHLIP uptake extends beyond hypoxic regions to stromal components, including metabolically active tumor-associated macrophages (TAM), which contribute to T-cell suppression [202,203,204]. Because acidosis promotes tumor progression, drug resistance, and immune dysfunction, imaging probes that map acidity may help predict and monitor responses to immunotherapy. Although pHLIPs have slower pharmacokinetics, the long half-life of 89Zr enables multi-day imaging, supporting their potential for clinical translation [201].
Beyond zirconium-labeled constructs, alternative radionuclides and chelation strategies have been explored to optimize pHLIP-based PET imaging. Both 18F and 64Cu can label NOTA- and NO2A-modified pHLIP peptides, with 18F offering broad availability and cost-effectiveness, while 64Cu provides a longer half-life, allowing for extended imaging windows. In 4T1 breast cancer models, negatively charged NOTA-conjugated variants clear rapidly via hepatobiliary and renal pathways, thereby limiting tumor retention. In contrast, neutrally charged NO2A-cys constructs exhibit slower blood clearance, improved tumor perfusion, and sustained trapping within acidic tumor regions. Among these, Var3 derivatives 64Cu-NO2A-cysVar3 and 18F-AlF-NO2A-cysVar3 exhibit the highest tumor uptake and tumor-to-background ratios across multiple models [205].
In summary, while FDG-PET does not directly measure pH, it provides a clinically accessible surrogate for regions of tumor acidosis in breast cancer, reflecting glycolytic activity and associated extracellular acidification. Together with pHLIP-based PET tracers that directly target acidic environments, these imaging approaches offer complementary strategies for visualizing tumor metabolism and pH, aiding in tumor characterization, prognostication, and potentially guiding therapy.

4. Discussion

This comprehensive review establishes tumor acidosis as a fundamental, targetable hallmark of breast cancer with profound implications for early detection. The inverted pH gradient, characterized by alkaline intracellular and acidic extracellular environments, is not merely a metabolic by product but an active driver of tumor progression, invasion, and treatment resistance. The central conclusion of this analysis is that pH-sensitive imaging represents a paradigm shift in breast cancer screening, moving beyond anatomical assessment to a functional and metabolic characterization.
The primary conclusion is that multiple imaging modalities have now matured to the point of providing reliable pH measurement [Table 1]. Techniques such as CEST MRI, particularly the exogenous acidoCEST approach, and advanced MRS methods (19F, 31P) offer clinically feasible pathways to quantify extracellular and intracellular pH with increasing precision. Hyperpolarized 13C MRI, despite its current technical complexity, provides unprecedented insight into real-time metabolic flux, with the lactate-to-pyruvate ratio emerging as a powerful predictive biomarker for treatment response. We included studies that demonstrated statistically significant pH-related findings, and readers are referred to the individual publications for detailed statistical methodologies and any correction procedures applied.
The relevance of these findings to the field is substantial and multifaceted. First, pH imaging addresses critical limitations of current screening and diagnostic techniques that rely on structural and morphological changes in tissue for differentiating malignancy from non-malignant processes, particularly in dense breasts, where structural modalities underperform. By detecting metabolic alterations that precede the formation of macroscopic tumors, these techniques can enable earlier cancer detection. Second, the ability to stratify tumors based on their pH profile and metabolic activity offers new avenues for personalized risk assessment and treatment selection, potentially identifying aggressive tumors that warrant more intensive management while sparing patients with indolent disease from overtreatment.
The translation of these technologies into routine clinical practice represents the next frontier. Although current pH Imaging techniques have been well studied and risk stratification and treatment monitoring, there remain limitations that preclude their use as broad screening tools for breast cancer screening. In particular, challenges in reproducibility, protocol standardization, cost, scanner availability, and validation across diverse patient populations must be addressed before large-scale clinical implementation is feasible. Importantly, emerging evidence linking pH-related imaging biomarkers such as hyperpolarized 13C lactate signals, elevated lactate on proton MRS, and higher endogenous CEST contrast to tumor stage and grade underscores the potential clinical value of these techniques once standardized and validated. Nevertheless, the convergence of growing biological understanding of tumor acidosis with rapid imaging innovation positions pH-based screening as a transformative approach. Future efforts should focus on integrating these functional techniques with existing structural modalities, developing standardized protocols, and validating pH thresholds that reliably predict clinical outcomes. With continued refinement and validation, pH-sensitive imaging holds the promise to revolutionize breast cancer detection by capturing disease at its earliest metabolic stages, ultimately improving personalized risk assessment, guiding timely intervention, and reducing mortality.

Author Contributions

Conceptualization, M.S. and D.K.; methodology, D.K.; writing—original draft preparation, D.K. and M.S.; writing—review and editing, S.G. and D.K.; supervision, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AMPKAMP-Activated Protein Kinase
AUCArea Under the Curve
DBTDigital Breast Tomosynthesis
DFODesferrioxamine
DMDigital Mammography
DQFDouble-Quantum Filtered
ECMExtracellular Matrix
EMTEpithelial-to-Mesenchymal Transition
ERKExtracellular Signal-Regulated Kinase
FDGFluorodeoxyglucose
FDG-PETFluorodeoxyglucose Positron Emission Tomography
gluCESTGlutamate Chemical Exchange Saturation Transfer
glycCESTGlycosaminoglycan Chemical Exchange Saturation Transfer
gagCESTGlycosaminoglycan Chemical Exchange Saturation Transfer
HER2Human Epidermal Growth Factor Receptor 2
HIF-1αHypoxia-Inducible Factor 1 alpha
LKB1Liver Kinase B1
LDHALactate Dehydrogenase A
MMP-9Matrix Metalloproteinase 9
NIRNear-Infrared
NOTA1,4,7-Triazacyclononane-1,4,7-triacetic Acid
NO2A1,4,7-Triazacyclononane-1,4-diacetic Acid
OCTOptical Coherence Tomography
PCPhosphocholine
pCRPathologic Complete Response
PEPhosphoethanolamine
PET-CTPositron Emission Tomography–Computed Tomography
pHLIPpH-Low Insertion Peptide
RSKRibosomal S6 Kinase
SLC16A1Solute Carrier Family 16 Member 1
SLC16A3Solute Carrier Family 16 Member 3
SLC4A7Solute Carrier Family 4 Member 7
SNARF-1Seminaphthorhodafluor-1
SNARF-5FSeminaphthorhodafluor-5F
SNRSignal-to-Noise Ratio
TAMTumor-Associated Macrophage
tChoTotal Choline
USPSTFUnited States Preventive Services Task Force

References

  1. Mahfuz Al Hasan, S.; Bennett, D.; Toriola, A. Screening programmes and breast cancer mortality: An observational study of 194 countries. Bull. World Health Organ. 2025, 103, 470–483. [Google Scholar] [CrossRef]
  2. Breast Cancer Statistics. How Common Is Breast Cancer? Available online: https://www.cancer.org/cancer/types/breast-cancer/about/how-common-is-breast-cancer.html (accessed on 29 October 2025).
  3. Kim, J.; Harper, A.; McCormack, V.; Sung, H.; Houssami, N.; Morgan, E.; Mutebi, M.; Garvey, G.; Soerjomataram, I.; Fidler-Benaoudia, M.M. Global patterns and trends in breast cancer incidence and mortality across 185 countries. Nat. Med. 2025, 31, 1154–1162. [Google Scholar] [CrossRef]
  4. US Preventive Services Task Force; Nicholson, W.K.; Silverstein, M.; Wong, J.B.; Barry, M.J.; Chelmow, D.; Coker, T.R.; Davis, E.M.; Jaén, C.R.; Krousel-Wood, M.; et al. Screening for Breast Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2024, 331, 1918–1930. [Google Scholar] [CrossRef]
  5. Turnbull, L.W. Dynamic contrast-enhanced MRI in the diagnosis and management of breast cancer. NMR Biomed. 2009, 22, 28–39. [Google Scholar] [CrossRef]
  6. Reig, B.; Lewin, A.A.; Du, L.; Heacock, L.; Toth, H.K.; Heller, S.L.; Gao, Y.; Moy, L. Breast MRI for Evaluation of Response to Neoadjuvant Therapy. Radiographics 2021, 41, 665–679. [Google Scholar] [CrossRef]
  7. Van Gils, C.H.; Otten, J.D.M.; Verbeek, A.L.M.; Hendriks, J.H.C.L. Mammographic breast density and risk of breast cancer: Masking bias or causality? Eur. J. Epidemiol. 1998, 14, 315–320. [Google Scholar] [CrossRef]
  8. Bodewes, F.T.H.; Van Asselt, A.A.; Dorrius, M.D.; Greuter, M.J.W.; De Bock, G.H. Mammographic breast density and the risk of breast cancer: A systematic review and meta-analysis. Breast 2022, 66, 62–68. [Google Scholar] [CrossRef] [PubMed]
  9. Trentham-Dietz, A.; Chapman, C.H.; Jayasekera, J.; Lowry, K.P.; Heckman-Stoddard, B.; Hampton, J.M.; Caswell-Jin, J.; Lu, Y.; Gangnon, R.E.; Sun, L.; et al. Breast Cancer Screening with Mammography: An Updated Decision Analysis for the U.S. Preventive Services Task Force; Agency for Healthcare Research and Quality: Rockville, MD, USA, 2024. Available online: http://www.ncbi.nlm.nih.gov/books/NBK603560/ (accessed on 29 October 2025).
  10. Trentham-Dietz, A.; Chapman, C.H.; Jayasekera, J.; Lowry, K.P.; Heckman-Stoddard, B.M.; Hampton, J.M.; Caswell-Jin, J.L.; Gangnon, R.E.; Lu, Y.; Huang, H.; et al. Collaborative Modeling to Compare Different Breast Cancer Screening Strategies: A Decision Analysis for the US Preventive Services Task Force. JAMA 2024, 331, 1947. [Google Scholar] [CrossRef]
  11. Nelson, H.D.; Cantor, A.; Humphrey, L.; Fu, R.; Pappas, M.; Daeges, M.; Griffin, J. Screening for Breast Cancer: A Systematic Review to Update the 2009 U.S. Preventive Services Task Force Recommendation; Agency for Healthcare Research and Quality: Rockville, MD, USA, 2016. Available online: http://www.ncbi.nlm.nih.gov/books/NBK343819/ (accessed on 29 October 2025).
  12. Henderson, J.T.; Webber, E.M.; Weyrich, M.; Miller, M.; Melnikow, J. Screening for Breast Cancer: A Comparative Effectiveness Review for the U.S. Preventive Services Task Force; Agency for Healthcare Research and Quality: Rockville, MD, USA, 2024. Available online: http://www.ncbi.nlm.nih.gov/books/NBK603789/ (accessed on 29 October 2025).
  13. Irvin, V.L.; Zhang, Z.; Simon, M.S.; Chlebowski, R.T.; Luoh, S.W.; Shadyab, A.H.; Krok-Schoen, J.L.; Tabung, F.K.; Qi, L.; Stefanick, M.L.; et al. Comparison of Mortality Among Participants of Women’s Health Initiative Trials with Screening-Detected Breast Cancers vs Interval Breast Cancers. JAMA Netw. Open 2020, 3, e207227. [Google Scholar] [CrossRef]
  14. Persi, E.; Duran-Frigola, M.; Damaghi, M.; Roush, W.R.; Aloy, P.; Cleveland, J.L.; Gillies, R.J.; Ruppin, E. Systems analysis of intracellular pH vulnerabilities for cancer therapy. Nat. Commun. 2018, 9, 2997. [Google Scholar] [CrossRef]
  15. Webb, B.A.; Chimenti, M.; Jacobson, M.P.; Barber, D.L. Dysregulated pH: A perfect storm for cancer progression. Nat. Rev. Cancer 2011, 11, 671–677. [Google Scholar] [CrossRef]
  16. Kato, Y.; Ozawa, S.; Miyamoto, C.; Maehata, Y.; Suzuki, A.; Maeda, T.; Baba, Y. Acidic extracellular microenvironment and cancer. Cancer Cell Int. 2013, 13, 89. [Google Scholar] [CrossRef]
  17. Kallinowski, F.; Schlenger, K.H.; Runkel, S.; Kloes, M.; Stohrer, M.; Okunieff, P.; Vaupel, P. Blood flow, metabolism, cellular microenvironment, and growth rate of human tumor xenografts. Cancer Res. 1989, 49, 3759–3764. [Google Scholar]
  18. Warburg, O.; Wind, F.; Negelein, E. The Metabolism Of Tumors In The Body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef] [PubMed]
  19. Lee, S.; Shanti, A. Effect of Exogenous pH on Cell Growth of Breast Cancer Cells. Int. J. Mol. Sci. 2021, 22, 9910. [Google Scholar] [CrossRef] [PubMed]
  20. Ward, C.; Meehan, J.; Gray, M.E.; Murray, A.F.; Argyle, D.J.; Kunkler, I.H.; Langdon, S.P. The impact of tumor pH on cancer progression: Strategies for clinical intervention. Explor. Target. Anti-Tumor Ther. 2020, 1, 71–100. [Google Scholar] [CrossRef]
  21. Estrella, V.; Chen, T.; Lloyd, M.; Wojtkowiak, J.; Cornnell, H.H.; Ibrahim-Hashim, A.; Bailey, K.; Balagurunathan, Y.; Rothberg, J.M.; Sloane, B.F.; et al. Acidity Generated by the Tumor Microenvironment Drives Local Invasion. Cancer Res. 2013, 73, 1524–1535. [Google Scholar] [CrossRef]
  22. Gillies, R.J.; Gatenby, R.A. Hypoxia and adaptive landscapes in the evolution of carcinogenesis. Cancer Metastasis Rev. 2007, 26, 311–317. [Google Scholar] [CrossRef]
  23. Morita, T.; Nagaki, T.; Fukuda, I.; Okumura, K. Clastogenicity of low pH to various cultured mammalian cells. Mutat. Res. 1992, 268, 297–305. [Google Scholar] [CrossRef] [PubMed]
  24. Shi, Q.; Le, X.; Wang, B.; Abbruzzese, J.L.; Xiong, Q.; He, Y.; Xie, K. Regulation of vascular endothelial growth factor expression by acidosis in human cancer cells. Oncogene 2001, 20, 3751–3756. [Google Scholar] [CrossRef]
  25. Bix, G.; Castello, R.; Burrows, M.; Zoeller, J.J.; Weech, M.; Iozzo, R.A.; Cardi, C.; Thakur, M.L.; Barker, C.A.; Camphausen, K.; et al. Endorepellin In Vivo: Targeting the Tumor Vasculature and Retarding Cancer Growth and Metabolism. JNCI J. Natl. Cancer Inst. 2006, 98, 1634–1646. [Google Scholar] [CrossRef]
  26. Wike-Hooley, J.L.; Haveman, J.; Reinhold, H.S. The relevance of tumor pH to the treatment of malignant disease. Radiother. Oncol. 1984, 2, 343–366. [Google Scholar] [CrossRef]
  27. Gillies, R.J.; Liu, Z.; Bhujwalla, Z. 31P-MRS measurements of extracellular pH of tumors using 3-aminopropylphosphonate. Am. J. Physiol. 1994, 267, C195–C203. [Google Scholar] [CrossRef]
  28. Tannock, I.F.; Rotin, D. Acid pH in tumors and its potential for therapeutic exploitation. Cancer Res. 1989, 49, 4373–4384. [Google Scholar] [PubMed]
  29. Swietach, P.; Boedtkjer, E.; Pedersen, S.F. How protons pave the way to aggressive cancers. Nat. Rev. Cancer 2023, 23, 825–841. [Google Scholar] [CrossRef] [PubMed]
  30. Koltai, T. The Ph paradigm in cancer. Eur. J. Clin. Nutr. 2020, 74, 14–19. [Google Scholar] [CrossRef]
  31. Michl, J.; Monterisi, S.; White, B.; Blaszczak, W.; Hulikova, A.; Abdullayeva, G.; Bridges, E.; Yin, Z.; Bodmer, W.F. Acid-adapted cancer cells alkalinize their cytoplasm by degrading the acid-loading membrane transporter anion exchanger 2, SLC4A2. Cell Rep. 2023, 42, 112601. [Google Scholar] [CrossRef] [PubMed]
  32. Boron, W.F. Regulation of intracellular pH. Adv. Physiol. Educ. 2004, 28, 160–179. [Google Scholar] [CrossRef]
  33. Chesler, M.; Nicholson, C. Regulation of intracellular pH in vertebrate central neurons. Brain Res. 1985, 325, 313–316. [Google Scholar] [CrossRef]
  34. Nishisho, T.; Hata, K.; Nakanishi, M.; Morita, Y.; Sun-Wada, G.-H.; Wada, Y.; Yasui, N.; Yoneda, T. The a3 isoform vacuolar type H+-ATPase promotes distant metastasis in the mouse B16 melanoma cells. Mol. Cancer Res. MCR 2011, 9, 845–855. [Google Scholar] [CrossRef] [PubMed]
  35. Pouysségur, J.; Franchi, A.; L’Allemain, G.; Paris, S. Cytoplasmic pH, a key determinant of growth factor-induced DNA synthesis in quiescent fibroblasts. FEBS Lett. 1985, 190, 115–119. [Google Scholar] [CrossRef]
  36. Moolenaar, W.H. Effects of growth factors on intracellular pH regulation. Annu. Rev. Physiol. 1986, 48, 363–376. [Google Scholar] [CrossRef]
  37. Kapus, A.; Romanek, R.; Qu, A.Y.; Rotstein, O.D.; Grinstein, S. A pH-sensitive and voltage-dependent proton conductance in the plasma membrane of macrophages. J. Gen. Physiol. 1993, 102, 729–760. [Google Scholar] [CrossRef]
  38. Denker, S.P.; Huang, D.C.; Orlowski, J.; Furthmayr, H.; Barber, D.L. Direct Binding of the Na–H Exchanger NHE1 to ERM Proteins Regulates the Cortical Cytoskeleton and Cell Shape Independently of H+ Translocation. Mol. Cell. 2000, 6, 1425–1436. [Google Scholar] [CrossRef]
  39. Lagadic-Gossmann, D.; Huc, L.; Lecureur, V. Alterations of intracellular pH homeostasis in apoptosis: Origins and roles. Cell Death Differ. 2004, 11, 953–961. [Google Scholar] [CrossRef]
  40. Matsuyama, S.; Llopis, J.; Deveraux, Q.L.; Tsien, R.Y.; Reed, J.C. Changes in intramitochondrial and cytosolic pH: Early events that modulate caspase activation during apoptosis. Nat. Cell Biol. 2000, 2, 318–325. [Google Scholar] [CrossRef]
  41. Kato, Y.; Ozawa, S.; Tsukuda, M.; Kubota, E.; Miyazaki, K.; St-Pierre, Y.; Hata, R. Acidic extracellular pH increases calcium influx-triggered phospholipase D activity along with acidic sphingomyelinase activation to induce matrix metalloproteinase-9 expression in mouse metastatic melanoma. FEBS J. 2007, 274, 3171–3183. [Google Scholar] [CrossRef]
  42. Kato, Y.; Nakayama, Y.; Umeda, M.; Miyazaki, K. Induction of 103-kDa gelatinase/type IV collagenase by acidic culture conditions in mouse metastatic melanoma cell lines. J. Biol. Chem. 1992, 267, 11424–11430. [Google Scholar] [CrossRef]
  43. Comito, G.; Iscaro, A.; Bacci, M.; Morandi, A.; Ippolito, L.; Parri, M.; Montagnani, I.; Raspollini, M.R.; Serni, S.; Simeoni, L.; et al. Lactate modulates CD4+ T-cell polarization and induces an immunosuppressive environment, which sustains prostate carcinoma progression via TLR8/miR21 axis. Oncogene 2019, 38, 3681–3695. [Google Scholar] [CrossRef]
  44. Certo, M.; Tsai, C.H.; Pucino, V.; Ho, P.C.; Mauro, C. Lactate modulation of immune responses in inflammatory versus tumor microenvironments. Nat. Rev. Immunol. 2021, 21, 151–161. [Google Scholar] [CrossRef]
  45. Calcinotto, A.; Filipazzi, P.; Grioni, M.; Iero, M.; De Milito, A.; Ricupito, A.; Cova, A.; Canese, R.; Jachetti, E.; Rossetti, M.; et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 2012, 72, 2746–2756. [Google Scholar] [CrossRef]
  46. Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef]
  47. Rolver, M.G.; Holland, L.K.K.; Ponniah, M.; Prasad, N.S.; Yao, J.; Schnipper, J.; Kramer, S.; Elingaard-Larsen, L.; Pedraz-Cuesta, E.; Liu, B.; et al. Chronic acidosis rewires cancer cell metabolism through PPARα signaling. Int. J. Cancer 2023, 152, 1668–1684. [Google Scholar] [CrossRef]
  48. Raghunand, N.; Gillies, R.J. pH and drug resistance in tumors. Drug Resist. Updat. 2000, 3, 39–47. [Google Scholar] [CrossRef]
  49. Gupta, S.; Farooque, A.; Adhikari, J.S.; Singh, S.; Dwarakanath, B.S. Enhancement of radiation and chemotherapeutic drug responses by 2-deoxy-D-glucose in animal tumors. J. Cancer Res. Ther. 2009, 5, S16–S20. [Google Scholar] [CrossRef]
  50. Pelicano, H.; Martin, D.S.; Xu, R.H.; Huang, P. Glycolysis inhibition for anticancer treatment. Oncogene 2006, 25, 4633–4646. [Google Scholar] [CrossRef]
  51. Hardie, D.G. AMP-activated/SNF1 protein kinases: Conserved guardians of cellular energy. Nat. Rev. Mol. Cell Biol. 2007, 8, 774–785. [Google Scholar] [CrossRef]
  52. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef]
  53. Toft, N.J.; Axelsen, T.V.; Pedersen, H.L.; Mele, M.; Burton, M.; Balling, E.; Johansen, T.; Thomassen, M.; Christiansen, P.M.; Boedtkjer, E. Acid-base transporters and pH dynamics in human breast carcinomas predict proliferative activity, metastasis, and survival. eLife 2021, 10, e68447. [Google Scholar] [CrossRef]
  54. Swallow, C.J.; Grinstein, S.; Rotstein, O.D. A vacuolar type H(+)-ATPase regulates cytoplasmic pH in murine macrophages. J. Biol. Chem. 1990, 265, 7645–7654. [Google Scholar] [CrossRef]
  55. Young, P.R.; Zygas, A.P. Secretion of lactic acid by peritoneal macrophages during extracellular phagocytosis. The possible role of local hyperacidity in inflammatory demyelination. J. Neuroimmunol. 1987, 15, 295–308. [Google Scholar] [CrossRef]
  56. Halestrap, A.P.; Wilson, M.C. The monocarboxylate transporter family--role and regulation. IUBMB Life 2012, 64, 109–119. [Google Scholar] [CrossRef]
  57. Doherty, J.R.; Yang, C.; Scott, K.E.; Cameron, M.D.; Fallahi, M.; Li, W.; Hall, M.A.; Amelio, A.L.; Mishra, J.K.; Li, F.; et al. Blocking lactate export by inhibiting the Myc target MCT1 Disables glycolysis and glutathione synthesis. Cancer Res. 2014, 74, 908–920. [Google Scholar] [CrossRef]
  58. Kumar, A.; Kant, S.; Singh, S.M. Targeting monocarboxylate transporter by α-cyano-4-hydroxycinnamate modulates apoptosis and cisplatin resistance of Colo205 cells: Implication of altered cell survival regulation. Apoptosis 2013, 18, 1574–1585. [Google Scholar] [CrossRef]
  59. Ullah, M.S.; Davies, A.J.; Halestrap, A.P. 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. [Google Scholar] [CrossRef]
  60. Xu, D.; Hemler, M.E. Metabolic activation-related CD147-CD98 complex. Mol. Cell. Proteomics MCP 2005, 4, 1061–1071. [Google Scholar] [CrossRef]
  61. Guo, H.; Li, R.; Zucker, S.; Toole, B.P. EMMPRIN (CD147), an inducer of matrix metalloproteinase synthesis, also binds interstitial collagenase to the tumor cell surface. Cancer Res. 2000, 60, 888–891. [Google Scholar]
  62. Martinez-Zaguilan, R.; Lynch, R.M.; Martinez, G.M.; Gillies, R.J. Vacuolar-type H(+)-ATPases are functionally expressed in plasma membranes of human tumor cells. Am. J. Physiol. 1993, 265, C1015–C1029. [Google Scholar] [CrossRef]
  63. Rochefort, H.; Liaudet, E.; Garcia, M. Alterations and role of human cathepsin D in cancer metastasis. Enzyme Protein 1996, 49, 106–116. [Google Scholar] [CrossRef]
  64. Montcourrier, P.; Mangeat, P.H.; Valembois, C.; Salazar, G.; Sahuquet, A.; Duperray, C.; Rochefort, H. Characterization of very acidic phagosomes in breast cancer cells and their association with invasion. J. Cell Sci. 1994, 107, 2381–2391. [Google Scholar] [CrossRef]
  65. Lauritzen, G.; Stock, C.M.; Lemaire, J.; Lund, S.F.; Jensen, M.F.; Damsgaard, B.; Petersen, K.S.; Wiwel, M.; Rønnov-Jessen, L.; Schwab, A.; et al. The Na+/H+ exchanger NHE1, but not the Na+, HCO3(-) cotransporter NBCn1, regulates motility of MCF7 breast cancer cells expressing constitutively active ErbB2. Cancer Lett. 2012, 317, 172–183. [Google Scholar] [CrossRef]
  66. Boedtkjer, E.; Moreira, J.M.A.; Mele, M.; Vahl, P.; Wielenga, V.T.; Christiansen, P.M.; Jensen, V.E.; Pedersen, S.F.; Aalkjaer, C. Contribution of Na+,HCO3(-)-cotransport to cellular pH control in human breast cancer: A role for the breast cancer susceptibility locus NBCn1 (SLC4A7). Int. J. Cancer 2013, 132, 1288–1299. [Google Scholar]
  67. Lee, S.; Mele, M.; Vahl, P.; Christiansen, P.M.; Jensen, V.E.D.; Boedtkjer, E. Na+,HCO3-cotransport is functionally upregulated during human breast carcinogenesis and required for the inverted pH gradient across the plasma membrane. Pflug. Arch. 2015, 467, 367–377. [Google Scholar] [CrossRef]
  68. Lee, S.; Axelsen, T.V.; Andersen, A.P.; Vahl, P.; Pedersen, S.F.; Boedtkjer, E. Disrupting Na+, HCO3-cotransporter NBCn1 (Slc4a7) delays murine breast cancer development. Oncogene 2016, 35, 2112–2122. [Google Scholar] [CrossRef]
  69. Lee, S.; Axelsen, T.V.; Jessen, N.; Pedersen, S.F.; Vahl, P.; Boedtkjer, E. Na+,HCO3-cotransporter NBCn1 (Slc4a7) accelerates ErbB2-induced breast cancer development and tumor growth in mice. Oncogene 2018, 37, 5569–5584. [Google Scholar] [CrossRef] [PubMed]
  70. Bernstein, B.W.; Painter, W.B.; Chen, H.; Minamide, L.S.; Abe, H.; Bamburg, J.R. Intracellular pH modulation of ADF/cofilin proteins. Cell Motil. Cytoskelet. 2000, 47, 319–336. [Google Scholar] [CrossRef]
  71. Kumar, A.P.; Quake, A.L.; Chang, M.K.X.; Zhou, T.; Lim, K.S.Y.; Singh, R.; Hewitt, R.E.; Salto-Tellez, M.; Pervaiz, S.; Clément, M.-V. Repression of NHE1 Expression by PPARγ Activation Is a Potential New Approach for Specific Inhibition of the Growth of Tumor Cells In Vitro and In Vivo. Cancer Res. 2009, 69, 8636–8644. [Google Scholar] [CrossRef]
  72. Miraglia, E.; Viarisio, D.; Riganti, C.; Costamagna, C.; Ghigo, D.; Bosia, A. Na+/H+ exchanger activity is increased in doxorubicin-resistant human colon cancer cells and its modulation modifies the sensitivity of the cells to doxorubicin. Int. J. Cancer 2005, 115, 924–929. [Google Scholar] [CrossRef]
  73. Sloth, R.A.; Axelsen, T.V.; Espejo, M.S.; Toft, N.J.; Voss, N.C.S.; Burton, M.; Thomassen, M.; Vahl, P.; Boedtkjer, E. Loss of RPTPγ primes breast tissue for acid extrusion, promotes malignant transformation and results in early tumor recurrence and shortened survival. Br. J. Cancer 2022, 127, 1226–1238. [Google Scholar] [CrossRef]
  74. Zhou, Y.; Skelton, L.A.; Xu, L.; Chandler, M.P.; Berthiaume, J.M.; Boron, W.F. Role of Receptor Protein Tyrosine Phosphatase γ in Sensing Extracellular CO2 and HCO3. J. Am. Soc. Nephrol. JASN 2016, 27, 2616–2621. [Google Scholar] [CrossRef]
  75. Boedtkjer, E.; Hansen, K.B.; Boedtkjer, D.M.B.; Aalkjaer, C.; Boron, W.F. Extracellular HCO3 is sensed by mouse cerebral arteries: Regulation of tone by receptor protein tyrosine phosphatase γ. J. Cereb. Blood. Flow. Metab. 2016, 36, 965–980. [Google Scholar] [CrossRef]
  76. Gottlieb, R.A.; Giesing, H.A.; Zhu, J.Y.; Engler, R.L.; Babior, B.M. Cell acidification in apoptosis: Granulocyte colony-stimulating factor delays programmed cell death in neutrophils by up-regulating the vacuolar H(+)-ATPase. Proc. Natl. Acad. Sci. USA 1995, 92, 5965–5968. [Google Scholar] [CrossRef]
  77. Khramtsov, V.; Grigor’ev, I.; Foster, M.; Lurie, D.J.; Nicholson, I. Biological applications of spin pH probes. Cell. Mol. Biol. 2000, 46, 1361–1374. [Google Scholar]
  78. Gillies, R.J.; Morse, D.L. In vivo magnetic resonance spectroscopy in cancer. Annu. Rev. Biomed. Eng. 2005, 7, 287–326. [Google Scholar] [CrossRef]
  79. Tognarelli, J.M.; Dawood, M.; Shariff, M.I.; Grover, V.P.; Crossey, M.M.; Cox, I.J.; Taylor-Robinson, S.D.; McPhail, M.J. Magnetic Resonance Spectroscopy: Principles and Techniques: Lessons for Clinicians. J. Clin. Exp. Hepatol. 2015, 5, 320–328. [Google Scholar] [CrossRef]
  80. Rhodes, C.J. Magnetic Resonance Spectroscopy. Sci. Prog. 2017, 100, 241–292. [Google Scholar] [CrossRef]
  81. Aime, S.; Botta, M.; Milone, L.; Terreno, E. Paramagnetic complexes as novel NMR pH indicators. Chem. Commun. 1996, 1265–1266. [Google Scholar] [CrossRef]
  82. Bitencourt, A.G.V.; Goldberg, J.; Pinker, K.; Thakur, S.B. Clinical applications of breast cancer metabolomics using high-resolution magic angle spinning proton magnetic resonance spectroscopy (HRMAS 1H MRS): Systematic scoping review. Metabolomics 2019, 15, 148. [Google Scholar] [CrossRef]
  83. Glunde, K.; Bhujwalla, Z.M.; Ronen, S.M. Choline metabolism in malignant transformation. Nat. Rev. Cancer 2011, 11, 835–848. [Google Scholar] [CrossRef]
  84. Sharma, U.; Jagannathan, N.R. In vivo MR spectroscopy for breast cancer diagnosis. BJR Open 2019, 1, 20180040. [Google Scholar] [CrossRef] [PubMed]
  85. Mellon, E.A.; Lee, S.; Pickup, S.; Kim, S.; Goldstein, S.C.; Floyd, T.F.; Poptani, H.; Delikatny, E.J.; Reddy, R.; Glickson, J.D. Detection of Lactate with a Hadamard Slice Selected, Selective Multiple Quantum Coherence, Chemical Shift Imaging Sequence (HDMD-SelMQC-CSI) on a clinical MRI scanner: Application to Tumors and Muscle Ischemia. Magn. Reson. Med. 2009, 62, 1404–1413. [Google Scholar] [CrossRef]
  86. Maudsley, A.A.; Darkazanli, A.; Alger, J.R.; Hall, L.O.; Schuff, N.; Studholme, C.; Yu, Y.; Ebel, A.; Frew, A.; Goldgof, D.; et al. Comprehensive processing, display and analysis for in vivo MR spectroscopic imaging. NMR Biomed. 2006, 19, 492–503. [Google Scholar] [CrossRef]
  87. He, Q.; Shungu, D.C.; van Zijl, P.C.; Bhujwalla, Z.M.; Glickson, J.D. Single-scan in vivo lactate editing with complete lipid and water suppression by selective multiple-quantum-coherence transfer (Sel-MQC) with application to tumors. J. Magn. Reson. B 1995, 106, 203–211. [Google Scholar] [CrossRef]
  88. Thakur, S.B.; Yaligar, J.; Koutcher, J.A. In vivo lactate signal enhancement using binomial spectral-selective pulses in selective MQ coherence (SS-SelMQC) spectroscopy. Magn. Reson. Med. 2009, 62, 591–598. [Google Scholar] [CrossRef]
  89. Payne, G.S.; Harris, L.M.; Cairns, G.S.; Messiou, C.; deSouza, N.M.; Macdonald, A.; Saran, F.; Leach, M.O. Validating a robust double-quantum-filtered (1) H MRS lactate measurement method in high-grade brain tumors. NMR Biomed. 2016, 29, 1420–1426. [Google Scholar] [CrossRef] [PubMed]
  90. Thakur, S.; Annarao, S.; Bokacheva, L.; Oh, J.H. Measurement of lactate concentrations in the breast mammary tumors using selective multiple quantum coherence editing sequence at 4.7 T. In Proceedings of the Joint Annual Meeting ISMRM-ESMRMB, Milan, Italy, 10–16 May 2014. [Google Scholar]
  91. Rizwan, A.; Serganova, I.; Khanin, R.; Karabeber, H.; Ni, X.; Thakur, S.; Zakian, K.L.; Blasberg, R.; Koutcher, J.A. Relationships between LDH-A, Lactate, and Metastases in 4T1 Breast Tumors. Clin. Cancer Res. 2013, 19, 5158–5169. [Google Scholar] [CrossRef]
  92. Serganova, I.; Rizwan, A.; Ni, X.; Thakur, S.B.; Vider, J.; Russell, J.; Blasberg, R.; Koutcher, J.A. Metabolic imaging: A link between lactate dehydrogenase A, lactate, and tumor phenotype. Clin. Cancer Res. 2011, 17, 6250–6261. [Google Scholar] [CrossRef]
  93. Cheung, S.M.; Husain, E.; Masannat, Y.; Miller, I.D.; Wahle, K.; Heys, S.D.; He, J. Lactate concentration in breast cancer using advanced magnetic resonance spectroscopy. Br. J. Cancer 2020, 123, 261–267. [Google Scholar] [CrossRef]
  94. García-Martín, M.L.; Hérigault, G.; Rémy, C.; Farion, R.; Ballesteros, P.; A Coles, J.; Cerdán, S.; Ziegler, A. Mapping extracellular pH in rat brain gliomas in vivo by 1H magnetic resonance spectroscopic imaging: Comparison with maps of metabolites. Cancer Res. 2001, 61, 6524–6531. [Google Scholar]
  95. van Sluis, R.; Bhujwalla, Z.M.; Raghunand, N.; Ballesteros, P.; Galons, J.-P.; Gillies, R.J.; Alvarez, J.; Cerdán, S. In vivo imaging of extracellular pH using 1H MRSI. Magn. Reson. Med. 1999, 41, 743–750. [Google Scholar] [CrossRef]
  96. Gil, M.S.; Cruz, F.; Cerdán, S.; Ballesteros, P. Imidazol-1-ylalkanoate esters and their corresponding acids. A novel series of extrinsic 1H NMR probes for intracellular pH. Bioorg. Med. Chem. Lett. 1992, 2, 1717–1722. [Google Scholar] [CrossRef]
  97. Gil, S.; Zaderenzo, P.; Cruz, F.; Cerdán, S.; Ballesteros, P. Imidazol-1-ylalkanoic acids as extrinsic 1H NMR probes for the determination of intracellular pH, extracellular pH and cell volume. Bioorg. Med. Chem. 1994, 2, 305–314. [Google Scholar] [CrossRef]
  98. Provent, P.; Benito, M.; Hiba, B.; Farion, R.; López-Larrubia, P.; Ballesteros, P.; Rémy, C.; Segebarth, C.; Cerdán, S.; Coles, J.A.; et al. Serial In vivo Spectroscopic Nuclear Magnetic Resonance Imaging of Lactate and Extracellular pH in Rat Gliomas Shows Redistribution of Protons Away from Sites of Glycolysis. Cancer Res. 2007, 67, 7638–7645. [Google Scholar] [CrossRef]
  99. Lee, S.H.; McIntyre, D.; Honess, D.; Hulikova, A.; Pacheco-Torres, J.; Cerdán, S.; Swietach, P.; Harris, A.L.; Griffiths, J.R. Carbonic anhydrase IX is a pH-stat that sets an acidic tumor extracellular pH in vivo. Br. J. Cancer 2018, 119, 622–630. [Google Scholar] [CrossRef]
  100. Bhujwalla, Z.M.; Artemov, D.; Ballesteros, P.; Cerdan, S.; Gillies, R.J.; Solaiyappan, M. Combined vascular and extracellular pH imaging of solid tumors. NMR Biomed. 2002, 15, 114–119. [Google Scholar] [CrossRef]
  101. Haselgrove, J.C.; Subramanian, V.H.; Leigh, J.S.; Gyulai, L.; Chance, B. In vivo one-dimensional imaging of phosphorus metabolites by phosphorus-31 nuclear magnetic resonance. Science 1983, 220, 1170–1173. [Google Scholar] [CrossRef]
  102. Podo, F. Tumor phospholipid metabolism. NMR Biomed. 1999, 12, 413–439. [Google Scholar]
  103. Glaholm, J.; Leach, M.; Collins, D.; Mansi, J.; Sharp, I.; Madden, A.; Smith, I.; Mccready, V. In-vivo 31P magnetic resonance spectroscopy for monitoring treatment response in breast cancer. Lancet 1989, 1, 1326–1327. [Google Scholar]
  104. Leach, M.O.; Verrill, M.; Glaholm, J.; Smith, T.A.D.; Collins, D.J.; Payne, G.S.; Sharp, J.C.; Ronen, S.M.; McCready, V.R.; Powles, T.J.; et al. Measurements of human breast cancer using magnetic resonance spectroscopy: A review of clinical measurements and a report of localized 31P measurements of response to treatment. NMR Biomed. 1998, 11, 314–340. [Google Scholar] [CrossRef]
  105. Arias-Mendoza, F.; Zakian, K.; Schwartz, A.; Howe, F.A.; Koutcher, J.A.; Leach, M.O.; Griffiths, J.R.; Heerschap, A.; Glickson, J.D.; Nelson, S.J.; et al. Methodological standardization for a multi-institutional in vivo trial of localized 31P MR spectroscopy in human cancer research. In vitro and normal volunteer studies. NMR Biomed. 2004, 17, 382–391. [Google Scholar] [CrossRef]
  106. Robey, I.F.; Baggett, B.K.; Kirkpatrick, N.D.; Roe, D.J.; Dosescu, J.; Sloane, B.F.; Hashim, A.I.; Morse, D.L.; Raghunand, N.; Gatenby, R.A.; et al. Bicarbonate Increases Tumor pH and Inhibits Spontaneous Metastases. Cancer Res. 2009, 69, 2260–2268. [Google Scholar] [CrossRef]
  107. Negendank, W. Studies of human tumors by MRS: A review. NMR Biomed. 1992, 5, 303–324. [Google Scholar] [CrossRef]
  108. Chen, L.Q.; Pagel, M.D. Evaluating pH in the Extracellular Tumor Microenvironment Using CEST MRI and Other Imaging Methods. Adv. Radiol. 2015, 2015, 1–25. [Google Scholar] [CrossRef] [PubMed]
  109. Sharma, U.; Jagannathan, N.R. Magnetic Resonance Imaging (MRI) and MR Spectroscopic Methods in Understanding Breast Cancer Biology and Metabolism. Metabolites 2022, 12, 295. [Google Scholar] [CrossRef] [PubMed]
  110. Rata, M.; Giles, S.L.; deSouza, N.M.; Leach, M.O.; Payne, G.S. Comparison of three reference methods for the measurement of intracellular pH using 31P MRS in healthy volunteers and patients with lymphoma. NMR Biomed. 2014, 27, 158–162. [Google Scholar] [CrossRef] [PubMed]
  111. Moon, R.B.; Richards, J.H. Determination of intracellular pH by 31P magnetic resonance. J. Biol. Chem. 1973, 248, 7276–7278. [Google Scholar] [CrossRef]
  112. Stubbs, M.; Bhujwalla, Z.M.; Tozer, G.M.; Rodrigues, L.M.; Maxwell, R.J.; Morgan, R.; Howe, F.A.; Griffiths, J.R. An assessment of 31P MRS as a method of measuring pH in rat tumors. NMR Biomed. 1992, 5, 351–359. [Google Scholar] [CrossRef]
  113. Ackerman, J.J.; Soto, G.E.; Spees, W.M.; Zhu, Z.; Evelhoch, J.L. The NMR chemical shift pH measurement revisited: Analysis of error and modeling of a pH dependent reference. Magn. Reson. Med. 1996, 36, 674–683. [Google Scholar] [CrossRef]
  114. Oberhaensli, R.D.; Hilton-Jones, D.; Bore, P.J.; Hands, L.J.; Rampling, R.P.; Radda, G.K. Biochemical investigation of human tumors in vivo with phosphorus-31 magnetic resonance spectroscopy. Lancet 1986, 328, 8–11. [Google Scholar] [CrossRef]
  115. Ng, T.C.; Grundfest, S.; Vijayakumar, S.; Baldwin, N.J.; Majors, A.W.; Karalis, I.; Meaney, T.F.; Shin, K.H.; Thomas, F.J.; Tubbs, R. Therapeutic response of breast carcinoma monitored by 31P MRS in situ. Magn. Reson. Med. 1989, 10, 125–134. [Google Scholar]
  116. Merchant, T.E.; Thelissen, G.R.; de Graaf, P.W.; Den Otter, W.; Glonek, T. Clinical magnetic resonance spectroscopy of human breast disease. Investig. Radiol. 1991, 26, 1053–1059. [Google Scholar] [CrossRef]
  117. Twelves, C.; Porter, D.; Lowry, M.; Dobbs, N.; Graves, P.; Smith, M.; Rubens, R.; Richards, M. Phosphorus-31 metabolism of post-menopausal breast cancer studied in vivo by magnetic resonance spectroscopy. Br. J. Cancer 1994, 69, 1151–1156. [Google Scholar] [CrossRef]
  118. van der Kemp, W.J.; Stehouwer, B.L.; Luijten, P.R.; van den Bosch, M.A.; Klomp, D.W. Detection of alterations in membrane metabolism during neoadjuvant chemotherapy in patients with breast cancer using phosphorus magnetic resonance spectroscopy at 7 Tesla. SpringerPlus 2014, 3, 634. [Google Scholar] [CrossRef] [PubMed]
  119. Li, H. A Non-invasive way to Detect Phospholipid Metabolism of Cancer: In vivo31P-MRS. J. Radiol. Radiat. Ther. 2024, 12, 1–7. [Google Scholar] [CrossRef]
  120. Ojugo, A.S.E.; McSheehy, P.M.J.; McIntyre, D.J.O.; McCoy, C.; Stubbs, M.; Leach, M.O.; Judson, I.R.; Griffiths, J.R. Measurement of the extracellular pH of solid tumors in mice by magnetic resonance spectroscopy: A comparison of exogenous (19)F and (31)P probes. NMR Biomed. 1999, 12, 495–504. [Google Scholar] [CrossRef]
  121. Gillies, R.J.; Raghunand, N.; Garcia-Martin, M.L.; Gatenby, R.A. pH imaging. A review of pH measurement methods and applications in cancers. IEEE Eng. Med. Biol. Mag. Q Mag. Eng. Med. Biol. Soc. 2004, 23, 57–64. [Google Scholar] [CrossRef]
  122. Deutsch, C.J.; Taylor, J.S. Intracellular pH as Measured by 19F NMRa. Ann. N. Y. Acad. Sci. 1987, 508, 33–47. [Google Scholar] [CrossRef]
  123. Chen, J.; Lanza, G.M.; Wickline, S.A. Quantitative magnetic resonance fluorine imaging: Today and tomorrow. WIREs Nanomed. Nanobiotechnol. 2010, 2, 431–440. [Google Scholar] [CrossRef]
  124. Prior, M.J.W.; Maxwell, R.J.; Griffiths, J.R. Fluorine-19F NMR Spectroscopy and Imaging In-Vivo. In In-Vivo Magnetic Resonance Spectroscopy III: In-Vivo MR Spectroscopy: Potential and Limitations; Rudin, M., Ed.; Springer: Berlin, Germany, 1992; pp. 101–130. [Google Scholar] [CrossRef]
  125. Mehta, V.D.; Kulkarni, P.V.; Mason, R.P.; Constantinescu, A.; Aravind, S.; Goomer, N.; Antich, P.P. 6-Fluoropyridoxol: A novel probe of cellular pH using 19F NMR spectroscopy. FEBS Lett. 1994, 349, 234–238. [Google Scholar]
  126. Deutsch, C.; Taylor, J.S.; Wilson, D.F. Regulation of intracellular pH by human peripheral blood lymphocytes as measured by 19F NMR. Proc. Natl. Acad. Sci. USA 1982, 79, 7944–7948. [Google Scholar] [CrossRef]
  127. Deutsch, C.; Taylor, J.S.; Price, M. pH homeostasis in human lymphocytes: Modulation by ions and mitogen. J. Cell Biol. 1984, 98, 885–893. [Google Scholar] [CrossRef]
  128. Taylor, J.S.; Deutsch, C.; McDonald, G.G.; Wilson, D.F. Measurement of transmembrane pH gradients in human erythrocytes using 19F NMR. Anal. Biochem. 1981, 114, 415–418. [Google Scholar] [CrossRef]
  129. Taylor, J.S.; Deutsch, C. Fluorinated alpha-methylamino acids as 19F NMR indicators of intracellular pH. Biophys. J. 1983, 43, 261–267. [Google Scholar] [CrossRef] [PubMed]
  130. McSheehy, P.; Seymour, M.; Ojugo, A.; Rodrigues, L.; Leach, M.; Judson, I.; Griffiths, J. A pharmacokinetic and pharmacodynamic study in vivo of human HT29 tumors using 19F and 31P magnetic resonance spectroscopy. Eur. J. Cancer 1997, 33, 2418–2427. [Google Scholar] [CrossRef]
  131. Peterson, K.L.; Srivastava, K.; Pierre, V.C. Fluorinated Paramagnetic Complexes: Sensitive and Responsive Probes for Magnetic Resonance Spectroscopy and Imaging. Front. Chem. 2018, 6, 160. [Google Scholar] [CrossRef]
  132. Mizukami, S. Development of Molecular Imaging Tools to Investigate Protein Functions by Chemical Probe Design. Chem. Pharm. Bull 2011, 59, 1435–1446. [Google Scholar] [CrossRef]
  133. Carril, M. Activatable probes for diagnosis and biomarker detection by MRI. J. Mater. Chem. B 2017, 5, 4332–4347. [Google Scholar] [CrossRef] [PubMed]
  134. Oishi, M.; Sumitani, S.; Nagasaki, Y. On–Off Regulation of 19F Magnetic Resonance Signals Based on pH-Sensitive PEGylated Nanogels for Potential Tumor-Specific Smart 19F MRI Probes. Bioconjug. Chem. 2007, 18, 1379–1382. [Google Scholar] [CrossRef]
  135. Yu, J.X.; Cui, W.; Bourke, V.A.; Mason, R.P. 6-Trifluoromethylpyridoxine: Novel 19F NMR pH Indicator for in Vivo Detection. J. Med. Chem. 2012, 55, 6814–6821. [Google Scholar] [CrossRef]
  136. Chen, S.; Yang, Y.; Li, H.; Zhou, X.; Liu, M. pH-Triggered Au-fluorescent mesoporous silica nanoparticles for 19F MR/fluorescent multimodal cancer cellular imaging. Chem. Commun. 2014, 50, 283–285. [Google Scholar] [CrossRef]
  137. Huang, X.; Huang, G.; Zhang, S.; Sagiyama, K.; Togao, O.; Ma, X.; Wang, Y.; Li, Y.; Soesbe, T.C.; Sumer, B.D.; et al. Multi-Chromatic pH-Activatable 19F-MRI Nanoprobes with Binary ON/OFF pH Transitions and Chemical-Shift Barcodes. Angew. Chem. Int. Ed. 2013, 52, 8074–8078. [Google Scholar]
  138. Li, Y.; Zhang, H.; Guo, C.; Hu, G.; Wang, L. Multiresponsive Nanoprobes for Turn-On Fluorescence/19F MRI Dual-Modal Imaging. Anal. Chem. 2020, 92, 11739–11746. [Google Scholar] [CrossRef]
  139. Zalewski, M.; Janasik, D.; Kapała, A.; Minoshima, M.; Sugihara, F.; Raj, W.; Pietrasik, J.; Kikuchi, K.; Krawczyk, T. pH-Sensitive Polymethacrylates as Potential Contrast Agents in 19F MRI. Macromol. Chem. Phys. 2022, 223, 2200027. [Google Scholar] [CrossRef]
  140. Janasik, D.; Jasiński, K.; Węglarz, W.P.; Nemec, I.; Jewula, P.; Krawczyk, T. Ratiometric pH-Responsive 19F Magnetic Resonance Imaging Contrast Agents Based on Hydrazone Switches. Anal. Chem. 2022, 94, 3427–3431. [Google Scholar] [CrossRef]
  141. Chen, L.; Jiang, Y.; Xiong, N.; Fan, Y.; Lin, H.; Gao, J. Sensitive Multichannel 19F Magnetic Resonance Imaging Enabled by Paramagnetic Fluorinated Ionic Liquid-Based Probes. ACS Nano 2025, 19, 9061–9069. [Google Scholar] [CrossRef] [PubMed]
  142. Deen, S.S.; Rooney, C.; Shinozaki, A.; McGing, J.; Grist, J.T.; Tyler, D.J.; Serrão, E.; Gallagher, F.A. Hyperpolarized Carbon 13 MRI: Clinical Applications and Future Directions in Oncology. Radiol. Imaging Cancer 2023, 5, e230005. [Google Scholar] [CrossRef]
  143. Gallagher, F.A.; Kettunen, M.I.; Brindle, K.M. Imaging pH with hyperpolarized 13C. NMR Biomed. 2011, 24, 1006–1015. [Google Scholar] [CrossRef]
  144. Woitek, R.; Brindle, K.M. Hyperpolarized Carbon-13 MRI in Breast Cancer. Diagnostics 2023, 13, 2311. [Google Scholar] [CrossRef]
  145. Gallagher, F.A.; Woitek, R.; McLean, M.A.; Gill, A.B.; Garcia, R.M.; Provenzano, E.; Riemer, F.; Kaggie, J.; Chhabra, A.; Ursprung, S.; et al. Imaging breast cancer using hyperpolarized carbon-13 MRI. Proc. Natl. Acad. Sci. USA 2020, 117, 2092–2098. [Google Scholar] [CrossRef]
  146. Woitek, R.; McLean, M.A.; Ursprung, S.; Rueda, O.M.; Garcia, R.M.; Locke, M.J.; Beer, L.; Baxter, G.; Rundo, L.; Provenzano, E.; et al. Hyperpolarized Carbon-13 MRI for Early Response Assessment of Neoadjuvant Chemotherapy in Breast Cancer Patients. Cancer Res. 2021, 81, 6004–6017. [Google Scholar] [CrossRef]
  147. Comment, A.; Merritt, M.E. Hyperpolarized magnetic resonance as a sensitive detector of metabolic function. Biochemistry 2014, 53, 7333–7357. [Google Scholar] [CrossRef] [PubMed]
  148. Ravoori, M.K.; Singh, S.P.; Lee, J.; Bankson, J.A.; Kundra, V. In Vivo Assessment of Ovarian Tumor Response to Tyrosine Kinase Inhibitor Pazopanib by Using Hyperpolarized 13C-Pyruvate MR Spectroscopy and 18F-FDG PET/CT Imaging in a Mouse Model. Radiology 2017, 285, 830–838. [Google Scholar] [CrossRef]
  149. Petersen, S.; Nagel, L.; Groß, P.R.; de Maissin, H.; Willing, R.; Heß, L.; Mitschke, J.; Klemm, N.; Treiber, J.; Müller, C.A.; et al. In Vivo Molecular Imaging of Breast Cancer Metabolic Heterogeneity Using [1-13C]Pyruvate-d3 Hyperpolarized By Reversible Exchange with Parahydrogen. Theranostics 2025, 15, 3714–3723. [Google Scholar] [CrossRef]
  150. Van Zijl, P.C.M.; Yadav, N.N. Chemical exchange saturation transfer (CEST): What is in a name and what isn’t? Magn Reson Med. 2011, 65, 927–948. [Google Scholar] [CrossRef]
  151. Cai, K.; Haris, M.; Singh, A.; Kogan, F.; Greenberg, J.H.; Hariharan, H.; Detre, J.A.; Reddy, R. Magnetic resonance imaging of glutamate. Nat. Med. 2012, 18, 302–306. [Google Scholar] [CrossRef]
  152. Sherry, A.D.; Woods, M. Chemical exchange saturation transfer contrast agents for magnetic resonance imaging. Annu. Rev. Biomed. Eng. 2008, 10, 391–411. [Google Scholar] [CrossRef]
  153. Song, X.; Airan, R.D.; Arifin, D.R.; Bar-Shir, A.; Kadayakkara, D.K.; Liu, G.; Gilad, A.A.; van Zijl, P.C.M.; McMahon, M.T.; Bulte, J.W.M. Label-free in vivo molecular imaging of underglycosylated mucin-1 expression in tumor cells. Nat. Commun. 2015, 6, 6719. [Google Scholar] [CrossRef]
  154. Sun, P.Z.; Lu, J.; Wu, Y.; Xiao, G.; Wu, R. Evaluation of the dependence of CEST-EPI measurement on repetition time, RF irradiation duty cycle and imaging flip angle for enhanced pH sensitivity. Phys. Med. Biol. 2013, 58, N229–N240. [Google Scholar] [CrossRef] [PubMed]
  155. Igarashi, T.; Kim, H.; Sun, P.Z. Detection of tissue pH with quantitative chemical exchange saturation transfer magnetic resonance imaging. NMR Biomed. 2023, 36, e4711. [Google Scholar] [CrossRef] [PubMed]
  156. Anemone, A.; Consolino, L.; Conti, L.; Irrera, P.; Hsu, M.Y.; Villano, D.; Dastrù, W.; Porporato, P.E.; Cavallo, F.; Longo, D.L. Tumor acidosis evaluated in vivo by MRI-CEST pH imaging reveals breast cancer metastatic potential. Br. J. Cancer 2020, 124, 207–216. [Google Scholar] [CrossRef]
  157. Corrado, A.; Lorito, N.; Anemone, A.; Carella, A.; Villano, D.; Pirotta, E.; Gammaraccio, F.; Subbiani, A.; Bacci, M.; Dastrù, W.; et al. In vivo imaging of the spatial heterogeneity of intratumoral acidosis (pH) as a marker of the metastatic phenotype in breast cancer. Breast Cancer Res. 2025, 27, 112. [Google Scholar] [CrossRef]
  158. Chan, K.W.Y.; Jiang, L.; Cheng, M.; Wijnen, J.P.; Liu, G.; Huang, P.; van Zijl, P.C.M.; McMahon, M.T.; Glunde, K. CEST-MRI detects metabolite levels altered by breast cancer cell aggressiveness and chemotherapy response. NMR Biomed. 2016, 29, 806–816. [Google Scholar] [CrossRef]
  159. Zhou, J.; Lal, B.; Wilson, D.A.; Laterra, J.; Van Zijl, P.C.M. Amide proton transfer (APT) contrast for imaging of brain tumors. Magn. Reson. Med. 2003, 50, 1120–1126. [Google Scholar] [CrossRef] [PubMed]
  160. Sun, P.Z.; Wang, E.; Cheung, J.S.; Zhang, X.; Benner, T.; Sorensen, A.G. Simulation and optimization of pulsed radio frequency irradiation scheme for chemical exchange saturation transfer (CEST) MRI—Demonstration of pH-weighted pulsed-amide proton CEST MRI in an animal model of acute cerebral ischemia. Magn. Reson. Med. 2011, 66, 1042–1048. [Google Scholar] [CrossRef]
  161. Sun, P.Z.; Wang, E.; Cheung, J.S. Imaging acute ischemic tissue acidosis with pH-sensitive endogenous amide proton transfer (APT) MRI—Correction of tissue relaxation and concomitant RF irradiation effects toward mapping quantitative cerebral tissue pH. NeuroImage 2012, 60, 1–6. [Google Scholar] [CrossRef] [PubMed]
  162. Zhou, J.; Payen, J.F.; Wilson, D.A.; Traystman, R.J.; van Zijl, P.C.M. Using the amide proton signals of intracellular proteins and peptides to detect pH effects in MRI. Nat. Med. 2003, 9, 1085–1090. [Google Scholar] [CrossRef]
  163. Zhou, J.; Heo, H.Y.; Knutsson, L.; van Zijl, P.C.M.; Jiang, S. APT-weighted MRI: Techniques, current neuro applications, and challenging issues. J. Magn. Reson. Imaging JMRI 2019, 50, 347–364. [Google Scholar] [CrossRef] [PubMed]
  164. Yan, K.; Fu, Z.; Yang, C.; Zhang, K.; Jiang, S.; Lee, D.-H.; Heo, H.-Y.; Zhang, Y.; Cole, R.N.; Van Eyk, J.E.; et al. Assessing Amide Proton Transfer (APT) MRI Contrast Origins in 9 L Gliosarcoma in the Rat Brain Using Proteomic Analysis. Mol. Imaging Biol. 2015, 17, 479–487. [Google Scholar] [CrossRef]
  165. Lee, D.; Heo, H.; Zhang, K.; Zhang, Y.; Jiang, S.; Zhao, X.; Zhou, J. Quantitative assessment of the effects of water proton concentration and water T1 changes on amide proton transfer (APT) and nuclear overhauser enhancement (NOE) MRI: The origin of the APT imaging signal in brain tumor. Magn. Reson. Med. 2016, 77, 855–863. [Google Scholar] [CrossRef]
  166. Xu, M.; Shan, D.; Zhang, R.; Li, J.; Guo, L.; Chen, X.; Qu, J. Differentiation of breast cancer subtypes and correlation with biological status using functional magnetic resonance imaging: Comparison with amide proton transfer-weighted imaging and diffusion-weighted imaging. Quant. Imaging Med. Surg. 2025, 15, 6102–6117. [Google Scholar] [CrossRef]
  167. Lee, R.C.; Boparai, M.S.; Duong, T.Q. Detection of breast cancer lesions using APT weighted MRI: A systematic review. J. Transl. Med. 2025, 23, 141. [Google Scholar] [CrossRef] [PubMed]
  168. Liu, Z.; Wen, J.; Wang, M.; Ren, Y.; Yang, Q.; Qian, L.; Luo, H.; Feng, S.; He, C.; Liu, X.; et al. Breast Amide Proton Transfer Imaging at 3 T: Diagnostic Performance and Association with Pathologic Characteristics. J. Magn. Reson. Imaging 2022, 57, 824–833. [Google Scholar] [CrossRef]
  169. Van Zijl, P.C.M.; Jones, C.K.; Ren, J.; Malloy, C.R.; Sherry, A.D. MRI detection of glycogen in vivo by using chemical exchange saturation transfer imaging (glycoCEST). Proc. Natl. Acad. Sci. USA 2007, 104, 4359–4364. [Google Scholar] [CrossRef]
  170. Ling, W.; Regatte, R.R.; Navon, G.; Jerschow, A. Assessment of glycosaminoglycan concentration in vivo by chemical exchange-dependent saturation transfer (gagCEST). Proc. Natl. Acad. Sci. USA 2008, 105, 2266–2270. [Google Scholar] [CrossRef]
  171. Zhou, R.; Bagga, P.; Nath, K.; Hariharan, H.; Mankoff, D.A.; Reddy, R. Glutamate-Weighted Chemical Exchange Saturation Transfer Magnetic Resonance Imaging Detects Glutaminase Inhibition in a Mouse Model of Triple-Negative Breast Cancer. Cancer Res. 2018, 78, 5521–5526. [Google Scholar] [CrossRef]
  172. Cember, A.T.J.; Nanga, R.P.R.; Reddy, R. Glutamate-weighted CEST (gluCEST) imaging for mapping neurometabolism: An update on the state of the art and emerging findings from in vivo applications. NMR Biomed. 2023, 36, e4780. [Google Scholar] [CrossRef] [PubMed]
  173. Ward, K.M.; Balaban, R.S. Determination of pH using water protons and chemical exchange dependent saturation transfer (CEST). Magn. Reson. Med. 2000, 44, 799–802. [Google Scholar] [CrossRef]
  174. Longo, D.L.; Dastrù, W.; Digilio, G.; Keupp, J.; Langereis, S.; Lanzardo, S.; Prestigio, S.; Steinbach, O.; Terreno, E.; Uggeri, F.; et al. Iopamidol as a responsive MRI-chemical exchange saturation transfer contrast agent for pH mapping of kidneys: In vivo studies in mice at 7 T. Magn. Reson. Med. 2010, 65, 202–211. [Google Scholar] [CrossRef]
  175. Longo, D.L.; Michelotti, F.; Consolino, L.; Bardini, P.; Digilio, G.; Xiao, G.; Sun, P.Z.; Aime, S. In Vitro and In Vivo Assessment of Nonionic Iodinated Radiographic Molecules as Chemical Exchange Saturation Transfer Magnetic Resonance Imaging Tumor Perfusion Agents. Investig. Radiol. 2016, 51, 155–162. [Google Scholar] [CrossRef]
  176. Chen, L.Q.; Howison, C.M.; Jeffery, J.J.; Robey, I.F.; Kuo, P.H.; Pagel, M.D. Evaluations of extracellular pH within in vivo tumors using acidoCEST MRI. Magn. Reson. Med. 2014, 72, 1408–1417. [Google Scholar] [CrossRef] [PubMed]
  177. Singh, A.; Stabinska, J.; Krishnamachary, B.; Sedaghat, F.; Nimmagadda, S.; Bulte, J.W.M.; Bhujwalla, Z.M.; McMahon, M.T. Glucose stimulated CEST MRI pHe mapping for improved differentiation of tumors with altered hypoxia inducible factor 1alpha expression. Sci. Rep. 2025, 15, 29129. [Google Scholar] [CrossRef]
  178. Chen, M.; Chen, C.; Shen, Z.; Zhang, X.; Chen, Y.; Lin, F.; Ma, X.; Zhuang, C.; Mao, Y.; Gan, H.; et al. Extracellular pH is a biomarker enabling detection of breast cancer and liver cancer using CEST MRI. Oncotarget 2017, 8, 45759–45767. [Google Scholar] [CrossRef]
  179. Moon, B.F.; Jones, K.M.; Chen, L.Q.; Liu, P.; Randtke, E.A.; Howison, C.M.; Pagel, M.D. A comparison of iopromide and iopamidol, two acidoCEST MRI contrast media that measure tumor extracellular pH. Contrast Media Mol. Imaging 2015, 10, 446–455. [Google Scholar] [CrossRef]
  180. Wu, R.; Longo, D.L.; Aime, S.; Sun, P.Z. Quantitative description of radiofrequency (RF) power-based ratiometric chemical exchange saturation transfer (CEST) pH imaging. NMR Biomed. 2015, 28, 555–565. [Google Scholar] [CrossRef]
  181. Wu, R.; Liu, C.; Liu, P.K.; Sun, P.Z. Improved measurement of labile proton concentration-weighted chemical exchange rate (kws) with experimental factor-compensated and T1-normalized quantitative chemical exchange saturation transfer (CEST) MRI. Contrast Media Mol. Imaging 2012, 7, 384–389. [Google Scholar] [CrossRef]
  182. Bakshi, S.F.; Guz, N.; Zakharchenko, A.; Deng, H.; Tumanov, A.V.; Woodworth, C.D.; Minko, S.; Kolpashchikov, D.M.; Katz, E. Nanoreactors based on DNAzyme-functionalized magnetic nanoparticles activated by magnetic field. Nanoscale 2017, 10, 1356–1365. [Google Scholar] [CrossRef]
  183. Ivanov, Y.D.; Pleshakova, T.; Malsagova, K.; Kozlov, A.; Kaysheva, A.; Shumov, I.; Galiullin, R.; Kurbatov, L.; Popov, V.; Naumova, O.; et al. Detection of marker miRNAs in plasma using SOI-NW biosensor. Sens. Actuators B Chem. 2018, 261, 566–571. [Google Scholar] [CrossRef]
  184. Dellian, M.; Helmlinger, G.; Yuan, F.; Jain, R. Fluorescence ratio imaging of interstitial pH in solid tumors: Effect of glucose on spatial and temporal gradients. Br. J. Cancer 1996, 74, 1206–1215. [Google Scholar] [CrossRef] [PubMed]
  185. Gatenby, R.A.; Gawlinski, E.T.; Gmitro, A.F.; Kaylor, B.; Gillies, R.J. Acid-Mediated Tumor Invasion: A Multidisciplinary Study. Cancer Res. 2006, 66, 5216–5223. [Google Scholar] [CrossRef] [PubMed]
  186. Hassan, M.; Riley, J.; Chernomordik, V.; Smith, P.; Pursley, R.; Lee, S.B.; Capala, J.; Gandjbakhche Amir, H. Fluorescence Lifetime Imaging System for In Vivo Studies. Mol. Imaging 2007, 6, 229–236. [Google Scholar] [CrossRef]
  187. Li, C.; Xia, J.; Wei, X.; Yan, H.; Si, Z.; Ju, S. pH-Activated Near-Infrared Fluorescence Nanoprobe Imaging Tumors by Sensing the Acidic Microenvironment. Adv. Funct. Mater. 2010, 20, 2222–2230. [Google Scholar] [CrossRef]
  188. Anemone, A.; Consolino, L.; Arena, F.; Capozza, M.; Longo, D.L. Imaging tumor acidosis: A survey of the available techniques for mapping in vivo tumor pH. Cancer Metastasis Rev. 2019, 38, 25–49. [Google Scholar] [CrossRef]
  189. Majd, S.M.; Salimi, A.; Ghasemi, F. An ultrasensitive detection of miRNA-155 in breast cancer via direct hybridization assay using two-dimensional molybdenum disulfide field-effect transistor biosensor. Biosens. Bioelectron. 2018, 105, 6–13. [Google Scholar] [CrossRef]
  190. Varghese, M.; Varghese, S.; Preethi, S. Revolutionizing medical imaging: A comprehensive review of optical coherence tomography (OCT). J. Opt. 2025, 54, 1178–1195. [Google Scholar] [CrossRef]
  191. Veselinovic, J.; Li, Z.; Daggumati, P.; Seker, E. Electrically Guided DNA Immobilization and Multiplexed DNA Detection with Nanoporous Gold Electrodes. Nanomaterials 2018, 8, 351. [Google Scholar] [CrossRef]
  192. Lin, Y.; Wu, T.Y.; Gmitro, A.F. Error analysis of ratiometric imaging of extracellular pH in a window chamber model. J. Biomed. Opt. 2012, 17, 046004. [Google Scholar] [CrossRef]
  193. Wang, Y.; Zhou, K.; Huang, G.; Hensley, C.; Huang, X.; Ma, X.; Zhao, T.; Sumer, B.D.; DeBerardinis, R.J.; Gao, J. A nanoparticle-based strategy for the imaging of a broad range of tumors by nonlinear amplification of microenvironment signals. Nat. Mater. 2014, 13, 204–212. [Google Scholar] [CrossRef]
  194. Ntziachristos, V.; Razansky, D. Molecular Imaging by Means of Multispectral Optoacoustic Tomography (MSOT). Chem. Rev. 2010, 110, 2783–2794. [Google Scholar] [CrossRef] [PubMed]
  195. Su, R.; Ermilov, S.; Liopo, A.; Oraevsky, A. Laser optoacoustic tomography: Towards new technology for biomedical diagnostics. Nucl. Instrum. Methods Phys. Res. Sect. A Accel. Spectrometers Detect. Assoc. Equip. 2013, 720, 58–61. [Google Scholar] [CrossRef]
  196. Stummer, W.; Pichlmeier, U.; Meinel, T.; Wiestler, O.D.; Zanella, F.; Reulen, H.J. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: A randomised controlled multicentre phase III trial. Lancet Oncol. 2006, 7, 392–401. [Google Scholar] [CrossRef]
  197. Chen, Q.; Liu, X.; Chen, J.; Zeng, J.; Cheng, Z.; Liu, Z. A Self-Assembled Albumin-Based Nanoprobe for In Vivo Ratiometric Photoacoustic pH Imaging. Adv. Mater. 2015, 27, 6820–6827. [Google Scholar] [CrossRef]
  198. Hofman, M.S.; Hicks, R.J. How We Read Oncologic FDG PET/CT. Cancer Imaging 2016, 16, 35. [Google Scholar] [CrossRef]
  199. Vanhove, K.; Thomeer, M.; Derveaux, E.; Shkedy, Z.; Owokotomo, O.E.; Adriaensens, P.; Mesotten, L. Correlations between the metabolic profile and 18F-FDG-Positron Emission Tomography-Computed Tomography parameters reveal the complexity of the metabolic reprogramming within lung cancer patients. Sci. Rep. 2019, 9, 16212. [Google Scholar] [CrossRef]
  200. Chardin, D.; Pourcher, T.; Gal, J.; Bailleux, C.; Guigonis, J.M.; Darcourt, J.; Arnould, L.; Humbert, O. Is “high FDG uptake” really synonymous with “hypermetabolism”? J. Nucl. Med. 2020, 61, 1214. Available online: https://jnm.snmjournals.org/content/61/supplement_1/1214 (accessed on 29 October 2025).
  201. Bauer, D.; Visca, H.; Weerakkody, A.; Carter, L.M.; Samuels, Z.; Kaminsky, S.; Andreev, O.A.; Reshetnyak, Y.K.; Lewis, J.S. PET Imaging of Acidic Tumor Environment with 89Zr-labeled pHLIP Probes. Front. Oncol. 2022, 12, 882541. [Google Scholar] [CrossRef] [PubMed]
  202. Rohani, N.; Hao, L.; Alexis, M.S.; Joughin, B.A.; Krismer, K.; Moufarrej, M.N.; Soltis, A.R.; Lauffenburger, D.A.; Yaffe, M.B.; Burge, C.B.; et al. Acidification of Tumor at Stromal Boundaries Drives Transcriptome Alterations Associated with Aggressive Phenotypes. Cancer Res. 2019, 79, 1952–1966. [Google Scholar] [CrossRef]
  203. Sahraei, M.; Chaube, B.; Liu, Y.; Sun, J.; Kaplan, A.; Price, N.L.; Ding, W.; Oyaghire, S.; García-Milian, R.; Mehta, S.; et al. Suppressing miR-21 activity in tumor-associated macrophages promotes an antitumor immune response. J. Clin. Investig. 2019, 129, 5518–5536. [Google Scholar] [CrossRef]
  204. Wu, H.; Estrella, V.; Beatty, M.; Abrahams, D.; El-Kenawi, A.; Russell, S.; Ibrahim-Hashim, A.; Longo, D.L.; Reshetnyak, Y.K.; Moshnikova, A.; et al. T-cells produce acidic niches in lymph nodes to suppress their own effector functions. Nat. Commun. 2020, 11, 4113. [Google Scholar] [CrossRef]
  205. Demoin, D.W.; Wyatt, L.C.; Edwards, K.J.; Abdel-Atti, D.; Sarparanta, M.; Pourat, J.; Longo, V.A.; Carlin, S.D.; Engelman, D.M.; Andreev, O.A.; et al. PET Imaging of Extracellular pH in Tumors with 64Cu- and 18F-Labeled pHLIP Peptides: A Structure–Activity Optimization Study. Bioconjug. Chem. 2016, 27, 2014–2023. [Google Scholar] [CrossRef] [PubMed]
  206. Javor, D.; Bennani-Baiti, B.I.; Clauser, P.; Kifjak, D.; Baltzer, P.A.T. Automated analysis of the total choline resonance peak in breast proton magnetic resonance spectroscopy. NMR Biomed. 2024, 37, e5054. [Google Scholar] [CrossRef]
  207. Fardanesh, R.; Marino, M.A.; Avendano, D.; Leithner, D.; Pinker, K.; Thakur, S.B. Proton MR spectroscopy in the breast: Technical innovations and clinical applications. J. Magn. Reson. Imaging 2019, 50, 1033–1046. [Google Scholar] [CrossRef]
  208. Cheung, S.M.; Husain, E.; Mallikourti, V.; Masannat, Y.; Heys, S.; He, J. Intra-tumoral lipid composition and lymphovascular invasion in breast cancer via non-invasive magnetic resonance spectroscopy. Eur. Radiol. 2021, 31, 3703–3711. [Google Scholar] [CrossRef] [PubMed]
  209. Van Der Kemp, W.J.M.; Van Der Velden, T.A.; Schmitz, A.M.; Gilhuijs, K.G.; Luijten, P.R.; Klomp, D.W.J.; Wijnen, J.P. Shortening of apparent transverse relaxation time of inorganic phosphate as a breast cancer biomarker. NMR Biomed. 2019, 32, e4011. [Google Scholar] [CrossRef]
  210. Jirak, D.; Galisova, A.; Kolouchova, K.; Babuka, D.; Hruby, M. Fluorine polymer probes for magnetic resonance imaging: Quo vadis? Magn. Reson. Mater. Phys. Biol. Med. 2019, 32, 173–185. [Google Scholar] [CrossRef]
  211. Jørgensen Sh Bøgh, N.; Hansen Ess Væggemose, M.; Wiggers, H.; Laustsen, C. Hyperpolarized MRI—An Update and Future Perspectives. Semin. Nucl. Med. 2022, 52, 374–381. [Google Scholar] [CrossRef]
  212. Vinogradov, E.; Keupp, J.; Dimitrov, I.E.; Seiler, S.; Pedrosa, I. CEST-MRI for body oncologic imaging: Are we there yet? NMR Biomed. 2023, 36, e4906. [Google Scholar] [CrossRef] [PubMed]
  213. Shimolina, L.; Potekhina, E.; Druzhkova, I.; Lukina, M.; Dudenkova, V.; Belousov, V.; Shcheslavskiy, V.; Zagaynova, E.; Shirmanova, M. Fluorescence lifetime-based pH mapping of tumors in vivo using genetically encoded sensor SypHerRed. Biophys. J. 2022, 121, 1156–1165. [Google Scholar] [CrossRef]
  214. Li, M.; Li, T.; Wu, F.; Ren, F.; Xue, S.; Li, C. Advanced NIR-II Fluorescence Imaging Technology for Precise Evaluation of Nanomedicine Delivery in Cancer Therapy. Chemosensors 2024, 12, 113. [Google Scholar] [CrossRef]
  215. Jo, J.; Lee, C.H.; Kopelman, R.; Wang, X. In vivo quantitative imaging of tumor pH by nanosonophore assisted multispectral photoacoustic imaging. Nat. Commun. 2017, 8, 471. [Google Scholar] [CrossRef]
  216. Wang, R.; Chen, H.; Fan, C. Impacts of time interval on 18F-FDG uptake for PET/CT in normal organs: A systematic review. Medicine 2018, 97, e13122. [Google Scholar] [CrossRef]
  217. Mann, R.M.; Cho, N.; Moy, L. Breast MRI: State of the Art. Radiology 2019, 292, 520–536. [Google Scholar] [CrossRef] [PubMed]
  218. Nguyen, F.T.; Zysk, A.M.; Chaney, E.J.; Kotynek, J.G.; Oliphant, U.J.; Bellafiore, F.J.; Rowland, K.M.; Johnson, P.A.; Boppart, S.A. Intraoperative Evaluation of Breast Tumor Margins with Optical Coherence Tomography. Cancer Res. 2009, 69, 8790–8796. [Google Scholar] [CrossRef]
Figure 1. Acidic tumor microenvironment in breast cancer, resulting from dysregulated metabolism. The Warburg effect leads to increased proton (H+) and lactate production, causing extracellular acidification and intracellular alkalinization. Key transporters and enzymes contributing to this pH dysregulation include monocarboxylate transporters (MCT1, MCT4), Na+/H+ exchanger (NHE1), H+-ATPase, Na+-HCO3 cotransporter (NBCn1), and carbonic anhydrases (CAIX, CAXII). pHe, extracellular pH; pHi, intracellular pH; TCA, tricarboxylic acid cycle; ATP, adenosine triphosphate; ADP, adenosine triphosphate.
Figure 1. Acidic tumor microenvironment in breast cancer, resulting from dysregulated metabolism. The Warburg effect leads to increased proton (H+) and lactate production, causing extracellular acidification and intracellular alkalinization. Key transporters and enzymes contributing to this pH dysregulation include monocarboxylate transporters (MCT1, MCT4), Na+/H+ exchanger (NHE1), H+-ATPase, Na+-HCO3 cotransporter (NBCn1), and carbonic anhydrases (CAIX, CAXII). pHe, extracellular pH; pHi, intracellular pH; TCA, tricarboxylic acid cycle; ATP, adenosine triphosphate; ADP, adenosine triphosphate.
Diagnostics 16 00076 g001
Figure 2. 31P-MRS estimates pH from the relative change in chemical shift of inorganic phosphate (Pi), which changes with pH, relative to phosphocreatine (PCr), which remains constant.
Figure 2. 31P-MRS estimates pH from the relative change in chemical shift of inorganic phosphate (Pi), which changes with pH, relative to phosphocreatine (PCr), which remains constant.
Diagnostics 16 00076 g002
Figure 3. Principle of Chemical exchange saturation transfer (CEST): the protons on the solute, once saturated, are exchangeable (blue) at a specific frequency, and this saturation is transferred to water following a period of tsat. RF, radiofrequency frequency; tsat, time required for saturation to be transferred to water.
Figure 3. Principle of Chemical exchange saturation transfer (CEST): the protons on the solute, once saturated, are exchangeable (blue) at a specific frequency, and this saturation is transferred to water following a period of tsat. RF, radiofrequency frequency; tsat, time required for saturation to be transferred to water.
Diagnostics 16 00076 g003
Table 1. Diagnostic Imaging techniques for pH detection for breast cancer screening.
Table 1. Diagnostic Imaging techniques for pH detection for breast cancer screening.
TechniquePrimary MeasureKey StrengthKey FindingMain
Limitation
Typical
Acquisition Times
Techniques to evaluate pH
1 H MRS (Conventional)Total Choline (tCho) concentrationHigh endogenous concentration; widely available on standard clinical MRI systems71–80% sensitivity/specificity for malignant lesion detectionIndirectly measures pH; lipid signal contamination5–10 min for single voxel spectroscopy [206]
10–15 min for multivoxel/2D [207]
1 H MRS (DQF)Lactate concentrationSpecific lactate detection by suppressing lipid signalsHigher lactate in grade III vs. grade II lesions; links to hypoxia50% inherent signal loss; challenging for small lesions10–12 min [93,208]
1 H MRS
(Exogenous Probes)
Chemical shift of probe’s H-2 protonRatiometric, concentration-independent pHe measurementSuccessful pHe mapping in preclinical models reveals acidic regionsSmall chemical shift range (~0.7 ppm); may alter native pHe~20–55 min [95,98]
31 P MRSChemical shift difference of inorganic phosphate compared to pH-independent phosphates IDirect measurement of intracellular pH; monitors energy metabolismResolves multiple pH compartments via Pi splittingLow endogenous concentration; poor spatial/temporal resolution~25 min [118,209]
19 F MRSChemical shift of exogenous 19F probeNegligible biological background; large chemical shift dispersionSuperior SNR vs. 31P MRS; enables specific pHe mappingRequires exogenous probes; limited clinical translation~5–20 min [210]
Hyperpolarized 13C MRILac/Pyr or H13CO3/13CO2 ratio>10,000× signal enhancement for real-time metabolic fluxLac/Pyr increase ≥20% predicts pCR post-treatmentExtreme cost/technical complexity; short signal lifetime~3 min [211]
CEST MRI
(Endogenous)
Amide proton transfer (APT) effectNo contrast agent needed; correlates with tumor aggressionHigh AUC (~0.96) for malignancy; tracks therapy responseConfounded by multiple factors; less sensitive in small lesions2–13 min [212]
CEST MRI
(Exogenous)
Chemical exchange of iodinated agentsRatiometric, concentration-independent pHe measurementRevealed “pH-neutral” tumors; quantitative pHe mappingLimited to acidic range (pH < 7.2); requires high field strength (e.g., 7 T)~30 min [156,176]
Fluorescence
Imaging
Fluorescence intensity ratioHigh sensitivity; real-time ratiometric quantificationDirect correlation between low pHe and high tumor invasionLimited to superficial tumors (<1 cm depth)Seconds to minutes [213,214]
Photoacoustic ImagingOptoacoustic signal ratioDeeper penetration than pure optical; combines optical/ultrasound91% sensitivity for lesions 1–2 mm; quantifies tumor pHNonlinear response curves; probe biocompatibility issuesSeconds [215]
Additional imaging modalities used in breast cancer that do not evaluate pH
FDG-PETGlucose uptake as a surrogate for glycolytic activityWidely available, non-invasive, clinically validated for staging and response assessmentHigher FDG uptake correlates with elevated glycolytic metabolites (lactate, L-acetylcarnitine), which are associated with extracellular acidificationIndirect measure of pH; cannot distinguish causes of high glucose flux (proliferation, inflammation, hypoxia)50–70 min [216]
89 Zr-pHLIP PETDirect binding of pHLIP peptides to acidic extracellular pHDirect visualization of acidic TME; identifies stromal acidity and TAM-rich regionsUniform tumor staining; highlights acidified zones beyond hypoxia, including immunosuppressive stromal regionsSlow pharmacokinetics; requires long-lived isotopesMultiday imaging (24–72 h post-injection [201])
18 F-AlF-NO2A-cysVar3/64Cu-NO2A-cysVar3 PETDirect pH-dependent peptide insertionPractical radiosynthesis (especially 18F-AlF); improved tumor retention; higher T:B ratiosNeutral NO2A-cysVar3 variants show sustained trapping in acidic TME and superior contrast across breast, prostate, melanoma, and brain tumorsSome variants show hepatobiliary clearance; optimization still ongoingMulti-day imaging [205] (24–72 h post-injection)
Dynamic Contrast-Enhanced MRIGadolinium uptake kineticsHighest reported sensitivity for breast cancer diagnosisClinical gold standard for lesion detection and characterizationRequires contrast injection; high cost; not pH-specific~5–7 min [217]
Optical
Coherence Tomography
Tissue microstructure changesHigh spatial resolution (~micrometers); non-invasive93% sensitivity/85% specificity for cancerous tissueDoes not directly measure pH; structural context only5 s per image [218]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kantamneni, D.; Gurbani, S.; Salvatore, M. The Role of Tumor pH in Breast Cancer Imaging: Biology, Diagnostic Applications, and Emerging Techniques. Diagnostics 2026, 16, 76. https://doi.org/10.3390/diagnostics16010076

AMA Style

Kantamneni D, Gurbani S, Salvatore M. The Role of Tumor pH in Breast Cancer Imaging: Biology, Diagnostic Applications, and Emerging Techniques. Diagnostics. 2026; 16(1):76. https://doi.org/10.3390/diagnostics16010076

Chicago/Turabian Style

Kantamneni, Dyutika, Saumya Gurbani, and Mary Salvatore. 2026. "The Role of Tumor pH in Breast Cancer Imaging: Biology, Diagnostic Applications, and Emerging Techniques" Diagnostics 16, no. 1: 76. https://doi.org/10.3390/diagnostics16010076

APA Style

Kantamneni, D., Gurbani, S., & Salvatore, M. (2026). The Role of Tumor pH in Breast Cancer Imaging: Biology, Diagnostic Applications, and Emerging Techniques. Diagnostics, 16(1), 76. https://doi.org/10.3390/diagnostics16010076

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop