Flow Cytometry, Ektacytometry, DNA Ploidy, and S-phase Fraction

Number: 0351

Table Of Contents

Policy
Applicable CPT / HCPCS / ICD-10 Codes
Background
References


Policy

Scope of Policy

This Clinical Policy Bulletin addresses flow cytometry, ektacytometry, DNA ploidy, and S-phase fraction.

  1. Medical Necessity

    1. Flow Cytometry for Cell Surface Markers

      Aetna considers flow cytometry for cell surface markers medically necessary for any of the following conditions:

      1. Abnormal tissue, bone marrow, or blood histology when the results are suspicious for lymphoma, leukemia, or myelodysplastic syndrome and where the physician must distinguish reactive from neoplastic conditions; or
      2. B-cell monitoring for immunosuppressive disorders; or
      3. Hairy cell monitoring; or
      4. Hereditary persistence of fetal hemoglobin (HPFH), in persons with unexplained increases in hemoglobin F; or
      5. Hereditary spherocytosis, in persons with Coombs' negative hemolytic anemia; or
      6. Immunophenotyping for leukemia, lymphoma, or myelodysplastic syndrome; or
      7. Mast cell disease; or
      8. Measurement of CD4/CD8 ratio from bronchiolar lavage fluid for diagnosis of sarcoidosis; or
      9. Multiple myeloma; or
      10. Myeloproliferative neoplasms, for workup of disease progression to advanced phase or transformation to AML; or
      11. Paroxysmal nocturnal hemoglobinuria; or
      12. Post-operative monitoring of members who have undergone organ transplantation; or
      13. Primary immunodeficiencies; or
      14. Sezary syndrome, diagnosis; or
      15. T-cell monitoring for HIV infection and AIDS.
    2. Ploidy and Cell Proliferation Activity: Indications

      A National Institutes of Health consensus development conference concluded that measurement of flow cytometry-derived DNA content (ploidy), or cell proliferative activity (S-phase fraction or % S-phase) is not indicated for prognostic or therapeutic purposes in the routine clinical management of cancers.  Therefore, Aetna considers flow cytometry-derived DNA content (ploidy), or cell proliferative activity (S-phase fraction or % S-phase) in any of the following localized cancers without metastatic disease medically necessary only when the obtained prognostic information will affect treatment decisions:

      1. Endometrial adenocarcinoma; or
      2. Gastric cancer; or
      3. Mediastinal neuroblastoma; or
      4. Medulloblastoma; or
      5. Ovarian carcinoma; or
      6. Partial hydatidiform mole; or
      7. Prostatic adenocarcinoma; or
      8. Renal cell adenocarcinoma; or
      9. Urinary bladder carcinoma.

      Note: This test is usually performed only once per tumor lifetime usually after a diagnosis has been made and before treatment is initiated.

    3. Ektacytometry

      Aetna considers ektacytometry medically necessary for the diagnosis of red blood cell (RBC) cytoskeleton and hydration disorders (e.g., hereditary spherocytosis, pyro-poikilocytosis, stomatocytosis, ovalocytosis, elliptocytosis and xerocytosis) when RBC morphology does not provide a clear diagnosis.

  2. Experimental and Investigational

    The following procedures are considered experimental and investigational because the effectiveness of these approaches has not been established:

    1. Aetna considers measurement of an RBC adhesion index (e.g., Hypoxic BioChip Adhesion or Normoxic BioChip Adhesion) experimental and investigational for all indications because the effectiveness of this approach has not been established.

    2. Aetna considers flow cytometry-derived DNA content (ploidy), or cell proliferative activity (S-phase fraction or % S-phase) experimental and investigational in any of the following cancers (not an all-inclusive list) because its effectiveness for these indications has not been established:

      1. Breast cancer; or
      2. Cervical cancer; or
      3. Colorectal cancer; or
      4. Non-small cell lung cancer; or
      5. Pediatric intracranial tumors; or
      6. Small cell lung cancer.

Table:

CPT Codes / HCPCS Codes / ICD-10 Codes

Code Code Description

Information in the [brackets] below has been added for clarification purposes.   Codes requiring a 7th character are represented by "+":

CPT codes covered if selection criteria are met:

86360 T cells; absolute CD4 and CD8 count, including ratio
88182 Flow cytometry, cell cycle or DNA analysis
88184 Flow cytometry, cell surface, cytoplasmic, or nuclear marker, technical component only; first marker
+ 88185     each additional marker (List separately in addition to code for first marker)
88187 Flow cytometry, interpretation; 2 to 8 markers
88188     9 to 15 markers
88189     16 or more markers

CPT codes not covered for indications listed in the CPB:

0303U Hematology, red blood cell (RBC) adhesion to endothelial/subendothelial adhesion molecules, functional assessment, whole blood, with algorithmic analysis and result reported as an RBC adhesion index; hypoxic
0304U Hematology, red blood cell (RBC) adhesion to endothelial/subendothelial adhesion molecules, functional assessment, whole blood, with algorithmic analysis and result reported as an RBC adhesion index; normoxic

ICD-10 codes covered if selection criteria are met:

B20 Human immunodeficiency virus [HIV] disease [T cell monitoring]
C16.0 - C16.9 Malignant neoplasm of stomach [gastric, localized without metastatic disease]
C38.1 - C38.2 Malignant neoplasm of anterior and posterior mediastinum [neuroblastoma, localized without metastatic disease]
C54.1 Malignant neoplasm of endometrium [localized without metastatic disease]
C56.1 - C56.9 Malignant neoplasm of ovary [localized without metastatic disease]
C57.4 Malignant neoplasm of uterine adnexa, unspecified [localized without metastatic disease]
C61 Malignant neoplasm of prostate
C64.1 - C64.9 Malignant neoplasm of unspecified kidney, except renal pelvis [localized without metastatic disease]
C67.0 - C67.9 Malignant neoplasm of bladder [localized without metastatic disease]
C71.0 - C71.9 Malignant neoplasm of brain [medulloblastoma in adults only]
C81.00 - C86.6
C88.4, C88.8 - C88.9
C90.00 - C94.42
C94.80 - C95.91
C96.0 - C96.4
C96.A - C96.9
Malignant neoplasm of lymphoid, hematopoietic and related tissue
D00.2 Carcinoma in situ of stomach [gastric]
D07.0 Carcinoma in situ of endometrium
D07.5 Carcinoma in situ of prostate
D09.0 Carcinoma in situ of bladder
D43.0 - D43.3 Neoplasm of uncertain behavior of brain and cranial nerves [intracranial in adults only]
D45 Polycythemia vera
D46.0 - D46.9 Myelodysplastic syndromes
D47.01 - D47.1
D47.3 - D47.9
Other neoplasms of uncertain behavior of lymphoid, hematopoietic and related tissue
D49.6 Neoplasm of unspecified behavior of brain [adults only]
D56.4 Hereditary persistence of fetal hemoglobin [HPFH]
D57.00 - D57.3
D57.80 - D57.819
Sickle-cell disorders
D58.0 Hereditary spherocytosis
D58.2 Other hemoglobinopathies
D59.5 - D59.8 Acquired hemolytic anemia
D75.81 Myelofibrosis
D80.0 - D81.2, D81.4
D81.89 - D82.1
D83.0 - D84.9
D89.810 - D89.9
Certain disorders involving the immune mechanism [B-cell monitoring for immunosuppressive disorders and primary immunodeficiencies]
D86.0 - D86.9 Sarcoidosis
O01.0 - O01.9 Hydatidiform mole
R89.7 Abnormal histological findings in specimens from other organs, systems and tissues
T86.00 - T86.99 Complications of transplanted organs and tissue [postoperative monitoring]
Z21 Asymptomatic human immunodeficiency virus [HIV] infection status [T cell monitoring]
Z94.0 - Z94.9, Z95.3 Transplanted organ and tissue status [postoperative monitoring]

ICD-10 code not covered for indications listed in the CPB:

C18.0 - C21.8 Malignant neoplasm of colon, rectosigmoid junction, rectum, anus and anal canal
C34.00 - C34.92 Malignant neoplasm of bronchus and lung [non-small cell lung cancer]
C50.011 - C50.929 Malignant neoplasm of breast
C53.0 - C53.9 Malignant neoplasm of cervix uteri
C71.0 - C71.9 Malignant neoplasm of brain [pediatric only]
D43.0 - D43.3 Neoplasm of uncertain behavior of brain and cranial nerves [pediatric intracranial only]
D49.6 Neoplasm of unspecified behavior of brain [pediatric only]

Ektacytometry:

CPT codes covered if selection criteria are met:

0305U Hematology, redbloodcell (RBC) functionality and deformity as a function of shear stress, whole blood, reported as a maximum elongation index

ICD-10 codes covered if selection criteria are met:

D58.0 Hereditary spherocytosis
D58.1 Hereditary elliptocytosis [Ovalocytosis]
D58.8 Other specified hereditary hemolytic anemias [Stomatocytosis, Xerocytosis]
R71.8 Other abnormality of red blood cells [pyro-poikilocytosis]

Background

Flow cytometry is an emerging technique, which involves the separation, classification and quantitation of cell types by:
  1. cell surface antigens (phenotype);
  2. DNA content (ploidy) (DNA index); and
  3. DNA flow cytometric proliferation analysis (S-phase fraction or % S-phase).
The basic technique involves passage of a monocellular stream of cells through a beam of laser light after cell surface antigens have been tagged with fluorescent monoclonal antibodies; complex computerized instruments are then used to sort normal from abnormal cells and also subgroups of the same cell type.  Data are most often collected as a bar-graph histogram, which is then displayed visually as a densitometer tracing of the bar graph; the concentration of cells in each bar appears as a separate peak for each cell category, with a peak height proportional to the number of cells in each bar of the bar graph.  For example, lymphocytes can be separated into B- and T-cell categories; the T-cells can be further phenotyped as helper/inducer, suppressor/cytotoxic, or natural killer cell types.

Using fluorescent dye that stains nucleic acids, flow cytometry methods have also been applied to measure nuclear deoxyribonucleic acid (DNA) content (ploidy) as a prognostic indicator of solid tumors based on the fact that malignant cells sometimes show abnormalities in total chromosome number and the frequency of these abnormalities generally increases with progression to higher-grade tumors.  In such testing, DNA diploid tumors are those in which a single peak containing an amount of DNA similar to normal control cells is generated by flow cytometry.  DNA aneuploid tumors have additional peaks on DNA histogram, presumably representing cells containing more or less nucleic acid than is found in 46 normal chromosomes.  A more quantitative method of expression is the DNA index (DI), which is the ratio of the mean tumor sample G0 /G1 DNA content of normal diploid reference cells.  The greater the deviation of the DI from 1, the more "aneuploid" the tumor.

Another type of measurement for DNA is the assessment of % S-phase or the S-phase fraction (SPF), the percentage or proportion of cells preparing for mitosis by their active doubling of DNA.  Since tumor cells tend to replicate more readily than normal cells, increased SPF activity can therefore raise the question of malignancy.  In many tumors, a high SPF correlates positively with poor differentiation, increasing tumor size, and degree of aggressiveness in tumor spread, which all have prognostic significance.

However, controversy exists concerning the use of DNA content (aneuploid versus diploid status) as an independent prognostic indicator.  Basic and clinical studies have reached different conclusions concerning its value.  While many of the earlier studies reported that diploid carcinomas had significantly or considerably better prognosis than aneuploid ones, some more recent studies do not confirm this or do not find that ploidy is a significant independent risk factor.  The results of studies on DNA content in different types of tumors have yielded varying results.  Aneuploidy, which is thought to be most often associated with malignancy, has been shown to occur in some non-neoplastic cell populations as part of the reaction to or regeneration after inflammation or tissue destruction and has also been reported in some benign tumors.  While groups of diploid patients have better prognoses than groups of non-diploid patients, ploidy status may have uncertain prognostic value in individual patients.  A small biopsy demonstrating diploid tumor may be missing a significant underlying non-diploid component.  Controversy remains as to the nature of the relationship between histologic grade and tumor stage with the degree of aneuploidy.  And, lastly, standards for tissue preparation and analysis to insure reproducibility are not yet established from lab to lab.

Increased S-phase activity is somewhat better accepted as an unfavorable prognostic sign.  Unfortunately, SPF is technically more difficult to measure accurately and its usefulness is limited by the lack of mutually accepted technical standards and methodologies between laboratories, including a uniform procedure for assaying cancers with multiple subpopulations of different kinetics.  Not all tumors with increased S-phase fraction are malignant; not all malignant tumors with increased S-phase fraction metastasize; and not all malignant tumors with relatively small S-phase fraction fail to metastasize.  Moreover, the S-phase peak is usually not large, even when considerable S-activity is occurring.  In DNA aneuploid tumors, the multiple peaks each represent a different cellular population; however, proliferation results are generally reported from only one peak, complicating the interpretation of the results.  Another problem of definition is that of high versus low S phase; these are frequently defined retrospectively or arbitrarily as being above or below the median for all samples assayed.  Such post hoc definitions may introduce bias and should in the future be replaced by laboratory-specific, prospectively determined, biologically relevant cutoffs.

The American Society of Clinical Oncology (2001) prepared evidence-based guidelines on the use of tumor markers in breast cancer and colorectal cancer, and reached the following conclusions.  Regarding DNA ploidy or flow cytometric proliferation analysis as a marker for colorectal cancer, ASCO concluded that "[p]resent data are insufficient to recommend DNA flow cytometrically-derived ploidy (DNA index) for the management of colorectal cancer."  Regarding the use of DNA flow cytometrically derived parameters as markers for breast cancer, ASCO concluded that:
  1. "[p]resent data are insufficient to recommend obtaining DNA flow cytometry-derived estimates of DNA content or S-phase in breast tissue"; and
  2. "DNA flow cytometry-derived ploidy are not recommended to be used to assign a patient to prognostic groupings.   There is insufficient evidence to recommend the use of S-phase determination for assigning patients to prognostic groupings."

In conclusion, DNA flow cytometry-derived estimates of DNA ploidy and S-phase proliferation have been correlated with cancer patient outcome in many studies.  However, the evidence is almost entirely from retrospective studies with multiple cut-off points for defining high- and low-risk populations.  The final value and role for DNA ploidy and S-phase fraction testing in current clinical practice remains to be finally determined.  The data are contradictory (hence the present controversy between the NIH and CMS's coverage policies) as to whether the prognostic information is independent of other recognized predictors, particularly stage, and further evaluation is required to determine if these techniques will provide useful prognostic information regarding individual patient survival, health outcome, and response to therapy.

The NCCN Biomarkers Compendium (2017) recognizes the use of flow cytometry for the following indications:

  • Acute lymphoblastic leukemia

    • Comprehensive flow cytometric immunophenotyping to include B, T and myeloid lineage markers;
    • Year 1 (every 1-2 months), bone marrow aspirate as indicated. If bone marrow aspirate is done: Flow cytometry with additional studies;
    • Minimal residual disease assessment - The most frequently employed methods for MRD assessment includes multicolor flow cytometry to detect abnormal immunophenotypes;

  • Acute myeloid leukemia: flow cytometry and IHC for evaluation of acute leukemia;
  • Chronic lymphocytic leukemia / small lymphocytic lymphoma: determination of CD38 and Zap 70 expression by flow cytometry, methylation or immunohistochemistry informative for prognostic or therapy determination;
  • Chronic myelogenous leukemia: flow cytometry as additional testing to determine cell lineage in advanced phase (accelerated phase or blast phase);
  • Multiple myeloma

    • Initial diagnostic workup: Unilateral bone marrow aspirate + biopsy, including bone marrow immunohistochemistry and/or bone marrow flow cytometry;
    • Smoldering (asymptomatic) clinical presentation - Multiparameter flow cytometry as clinically indicated for follow up/surveillance;

  • Myelodysplastic syndromes

    • Additional testing: Consider flow cytometry (FCM) to evaluate for PNH clone;
    • Helpful in some clinical situations:

      • Consider flow cytometry (FCM) for MDS diagnostic aid;
      • Consider flow cytometry (FCM) to assess possible large granular lymphocytic (LGL) disease;

  • Myeloproliferative neoplasms: advanced phase/AML workup;
  • Non-hodgkin lymphoma: diagnosis.

Habermehl et al (2004) examined flow-cytometric DNA values of pediatric intra-cranial tumors and attempted to establish DNA analysis as a potential prognostic parameter.  A total of 29 brain tumor specimens from 26 pediatric patients were cryo-preserved within a 3-year period.  The DNA content was measured by flow cytometry.  Six of the tumor specimens had aneuploid DNA patterns.  The median of the proliferation index was lower in the survivor group compared with the non-survivor group (36.4 and 47.5 %, respectively).  Ten of the 26 patients are still alive, 8 were lost to follow-up, and 8 died.  Flow-cytometric DNA analysis may be a helpful tool for examining brain tumors in children.  The small size of this study could not establish flow cytometry as a definite prognostic factor.  Further prospective multi-center studies are needed to assess the prognostic significance of flow-cytometric DNA analysis.

Gastric cancer is still a common cause of cancer-related deaths worldwide, despite improved diagnostic and therapeutic implications.  Hence, early diagnosis has critical importance.  Flow cytometry reveals rapid and reproducible quantification of nuclear DNA content of disaggregated tissues and assessment of its significance in various malignant and pre-cancerous lesions.  In a multi-center study, Yasa and colleagues (2005) examined 121 patients with gastric cancer, chronic atrophic gastritis, gastric polyps, intestinal metaplasia, and gastric dysplasia as well as 36 healthy controls.  Flow cytometric measurements of DNA ploidy, total S-phase, G2M-phase and proliferative indexes were analyzed on fresh gastric biopsy specimens obtained by gastroscopy.  DNA aneuploidy was present in 43.8 % of the gastric cancers (p < 0.05).  These investigators found a DNA aneuploidy rate of 15.4 % in chronic atrophic gastritis, 15.4 % in intestinal metaplasia and 25 % in epithelial dysplasia.  One of 9 polyps had aneuploidy.  None of the normal gastric mucosa samples showed aneuploidy.  The controls had lower rates of total S-phase and proliferative indexes (p < 0.05).  The authors concluded that DNA flow cytometry may be offered as an objective diagnostic tool for early detection of malignant transformation in gastric lesions.  Moreover, Russo et al (2001) who stated that DNA ploidy and SPF, when associated with clinico-pathological staging, might be useful for the identification of gastric cancer patients who have different risks for death or relapse of disease.  In addition, Jiao et al (2004) noted that SPF may be a more useful indicator of aggressive behavior in gastric cancers than DNA aneuploidy.

The American Society of Clinical Oncology's update of recommendations for the use of tumor markers in breast cancer (Harris et al, 2007) noted that there is insufficient evidence to support routine use in clinical practice of DNA/ploidy by flow cytometry.  ASCO's updated recommendations on the use of tumor markers in colorectal cancer state that neither flow-cytometrically derived DNA ploidy nor DNA flow cytometric proliferation analysis (% S phase) should be used to determine prognosis of early-stage colorectal cancer (Locker et al, 2006).

Wolfson et al (2008) examined possible associations between measurements of DI, SPF, and tumor heterogeneity (TH) using flow cytometry and overall survival (OS) for patients with invasive cervical carcinoma treated with definitive irradiation.  A total of 57 patients with International Federation of Obstetrics and Gynecology Stages IB(2) through IVB cervical carcinomas treated with definitive radiotherapy with or without concurrent chemotherapy were enrolled into this registry study that involved flow cytometric analysis of fresh tissue from each cervical cancer obtained by pre-treatment biopsy.  These specimens were evaluated for DNA aneuploidy (DI less than or equal to 1.5 versus greater than 1.5), SPF (less than or equal to 15 % versus greater than 15 %), and TH (uniploid versus multiploid).  In these analyses, 27 of the patients were treated in Radiation Therapy Oncology Group protocol 9001, and an additional 30 were offered chemoradiation at a single institution.  Forty-one patients had DI less than or equal to 1.5 and 16 DI greater than 1.5.  Twenty-nine patients had SPF less than or equal to 15 %, 26 greater than 15 %, and 2 had no determinable SPF.  Forty-three patients had uniploid and 14 multiploid tumors.  The 4-year estimated OS rate for the entire study cohort was 62 % (95 % confidence interval: 48 % to 74 %).  With a median follow-up of 3.7 years, there were no observable associations by univariate analysis for DI, SPF, or TH concerning patient survival.  The authors concluded that there were no statistically significant associations among DI, SPF, or TH and patient outcome.  They stated that additional studies are needed to identify tumor biomarkers that could predict patients at risk for disseminated disease.  Furthermore, Dabic et al (2008) stated that clinical stage and architectural grade are significant predictors for survival of patients with cervical adenocarcinoma.  Status of HPV infection, flow cytometric parameters, nuclear grade and menstrual status do not predict patient survival.

Suehiro and colleagues (2008) stated that many investigators have reported that aneuploidy detected by flow cytometry is a useful prognostic marker in patients with endometrial cancer.  Laser scanning cytometry (LSC) is a technology similar to flow cytometry but is more feasible for clinical laboratory use.  These investigators evaluated the usefulness of DNA ploidy detected by LSC as a prognostic marker in patients with endometrial cancer and examined genetic and epigenetic factors related to aneuploidy.  Endometrial cancer specimens from 106 patients were evaluated.  The methylation status of CDH13, Rassf1, SFRP1, SFRP2, SFRP4, SFRP5, p16, hMLH1, MGMT, APC, ATM, and WIF1 and mutations in the p53 and CDC4 genes were investigated.  Laser scanning cytometry was performed to determine DNA ploidy.  Fluorescence in situ hybridization was done with chromosome-specific centromeric probes to assess chromosomal instability.  Uni-variate and multi-variate analyses revealed that p53 mutation and lack of CDH13 hypermethylation associated positively with aneuploidy.  Uni-variate analysis showed that aneuploidy, chromosomal instability, and lack of CDH13 hypermethylation as well as surgical stage were significantly predictive of death from endometrial cancer.  Furthermore, multi-variate analysis revealed that stage in combination with either DNA aneuploidy or lack of CDH13 hypermethylation was an independent prognostic factor.  The authors concluded that these findings suggested that analysis of DNA ploidy and methylation status of CDH13 may help predict clinical outcome in patients with endometrial cancer.  Moreover, they stated that prospective randomized trials are needed to confirm the validity of an individualized approach, including determination of tumor ploidy and methylation status of CDH13, to management of endometrial cancer patients.

Ludovini et al (2008) assessed the relationship between a panel of biological markers (p53, Bcl-2, HER-2, Ki67, DNA ploidy and S-phase fraction) and clinical-pathological parameters and its impact on outcome in non-small cell lung cancer (NSCLC).  Tumor tissue specimens obtained following surgical resection were collected from consecutive patients with NSCLC.  These researchers used an immunocytochemical technique for p53, Bcl-2, HER-2 and Ki67 analysis in fine-needle aspirates obtained from surgical samples that were also evaluated by flow cytometric DNA analysis using a FACScan flow cytometer.  From April 2000 to December 2005, a total of 136 patients with radically resected NSCLC were recruited.  Median age was 66 years (range of 31 to 84 years), male/female ratio 117/19, ECOG performance status 0/1 127/4, stage I/II/III 76/25/35, squamous/adenocarcinoma/large-cell/mixed histology 62/49/17/8, smokers yes/no 121/11.  Positivity of p53, Bcl-2, HER-2 and Ki67 was detected in 51.4 %, 27.9 %, 25.0 % and 55.8 % of the samples, respectively; 82.9 % of the cases revealed aneuploid DNA histograms and 56.7 % presented an S-phase fraction of more than 12 %.  Statistically significant associations between high Ki67 and poorly differentiated tumors (p = 0.016) and a smoking history (p = 0.053); p53 positivity and high Ki67 (p = 0.002); HER-2 positivity and adenocarcinoma subtype (p = 0.015) and presence of lymph node involvement (p = 0.006); and Bcl-2 positivity and squamous cell carcinoma subtype (p = 0.058) were observed.  At uni-variate analysis, high Ki67 proved to be the only marker associated with disease-free survival (p = 0.047).  After adjusting for stage, none of the examined immunocytochemical markers emerged as an independent factor for disease-free and overall survival; only pathological stage was identified as an independent prognostic factor for disease-free survival (p = 0.0001) and overall survival (p = 0.0001).  In the group of 76 patients classified as TNM stage I, high Ki67 was the only marker associated with recurrence of disease (p = 0.05).  The authors concluded that these findings do not support a relevant prognostic role of immunocytochemical markers in NSCLC, even if the Ki67 index might have particular relevance to identify patients with more aggressive tumors who are at high risk for disease relapse.

The American Thoracic Society’s "Statement on Sarcoidosis" (1999) stated that "In some instances, bronchoalvcolar lavage (BAL) and studies on lymphocyte subpopulations are helpful.  According to Costabel, a CD41CD8 ratio greater than 3.5 has a sensitivity of 53 %, a specificity of 94 %, a positive-predictive value of 76 % and a negative-predictive value of 85 %.  In other words, a CD4/CD8 ratio greater than 3.5 provides a diagnosis of sarcoidosis with a specificity of 94 % even if the TLB has not been diagnostic".

Furthermore, an UpToDate review on "Clinical manifestations and diagnosis of pulmonary sarcoidosis" (King, 2015) states that "Bronchoalveolar lavage -- BAL can be used as an adjunctive measure to support the diagnosis of sarcoidosis by demonstrating a reduced number of CD8 cells, an elevated CD4 to CD8 ratio, and an increased amount of activated T cells, CD4 cells, immunoglobulins, and IgG-secreting cells.  BAL is also used to exclude infections and malignancy as alternative diagnoses".

National Comprehensive Cancer Network's guidelines on myeloproliferative neoplasms (NCCN, 2017) states that bone marrow aspirate and biopsy with reticulin stain and bone marrow cytogenetics, flow cytometry, and molecular testing for AML-associated mutations is recommended as part of initial workup of disease progression to advanced stage or transformation to AML.

Small Cell Lung Cancer

An UpToDate review on "Overview of the initial evaluation, treatment and prognosis of lung cancer" (Midthun, 2016) does not mention flow cytometry as a management tool.

Furthermore, National Comprehensive Cancer Network’ clinical practice guideline on "Small cell lung cancer" (Version 1.2016) does not mention flow cytometry as a management tool.

Osmotic Gradient Ektacytometry

Osmotic gradient ektacytometry (OGE; osmoscan) is part of the laboratory diagnosis process of hereditary spherocytosis (HS) and other red blood cell (RBC) membrane disorders.

Parrow and associates (2018) noted that decreased RBC deformability is characteristic of several disorders.  In some cases, the extent of defective deformability could predict severity of disease or occurrence of serious complications.  Ektacytometry uses laser diffraction viscometry to measure the deformability of RBCs subject to either increasing shear stress or an osmotic gradient at a constant value of applied shear stress.  However, direct deformability measurements are difficult to interpret when measuring heterogenous blood that is characterized by the presence of both rigid as well as deformable RBCs.  This is due to the inability of rigid cells to properly align in response to shear stress and results in a distorted diffraction pattern marked by an exaggerated decrease in apparent deformability.  Measurement of the degree of distortion provides an indicator of the heterogeneity of the erythrocytes in blood.  In sickle cell anemia, this is correlated with the percentage of rigid cells, which reflects the hemoglobin (Hb) concentration and Hb composition of RBCs.  In addition to measuring deformability, OGE provides information regarding the osmotic fragility and hydration status of RBCs.  These parameters also reflect the Hb composition of RBCs from sickle cell patients.  Ektacytometry measures deformability in populations of RBCs and does not, therefore, provide information on the deformability or mechanical properties of individual RBCs.  The authors concluded that these techniques may be useful for monitoring temporal changes, as well as disease progression and response to therapeutic intervention in several disorders; and sickle cell anemia is one well-characterized example.  Other potential disorders where measurements of RBC deformability and/or heterogeneity are of interest include blood storage, diabetes, Plasmodium infection, iron deficiency, and hemolytic anemias due to membrane defects.

Llaudet-Planas and colleagues (2018) stated that new generation OGE has become a powerful procedure for measuring RBC deformability and thus for the diagnosis of RBC membrane disorders.  These investigators provided further support to the usefulness of OGE for the differential diagnosis of HS by measuring the optimal cut-off values of the parameters provided by this technique.  A total of 65 cases of HS, 7 hereditary elliptocytosis (HE), 3 hereditary xerocytosis (HX), and 171 normal controls were analyzed with OGE in addition to the routine RBC laboratory techniques.  The most robust OGE parameters for HS diagnosis were determined using receiver operating characteristic (ROC) curve analysis.  The best diagnostic criteria for HS were the combination of decreased minimal elongation index up to 3 % and increased minimal osmolality point up to 5.2 % when compared to the mean of controls.  Using this established criterion, OGE reported a sensitivity of 93.85 % and a specificity of 98.38 % for the diagnosis of HS.  The authors concluded that OGE is an effective diagnostic test for HS and enables its differential diagnosis with other RBC membrane diseases based on specific pathology profiles.

Huisjes and associates (2020) noted that HS originates from defective anchoring of the cytoskeletal network to the trans-membrane protein complexes of RBCs.  Red cells in HS are characterized by membrane instability and reduced deformability and there is marked heterogeneity in disease severity among patients.  To unravel this variability in the severity of HS, these researchers analyzed blood samples from 21 HS patients with defects in ankyrin, band 3, α-spectrin or β-spectrin by means of red cell indices, eosin-5-maleimide binding, microscopy, the osmotic fragility test, Percoll density gradients, vesiculation and OGE to evaluate cell membrane stability, cellular density and deformability.  Reticulocyte counts, CD71 abundance, band 4.1 a:b ratio, and glycated Hb were used as markers of RBC turnover.  These investigators observed that patients with moderate/severe HS have short-living RBCs of low density and abnormally high intercellular heterogeneity.  These cells showed a prominent decrease in membrane stability and deformability and, as a consequence, were quickly removed from the circulation by the spleen.  In contrast, in mild HS less pronounced reduction in deformability resulted in prolonged RBC lifespan and, hence, cells were subjected to progressive loss of membrane.  RBCs from patients with mild HS thus become denser before they were taken up by the spleen.  The authors concluded that RBC membrane loss, cellular heterogeneity and density were strong markers of clinical severity in patients with HS.

Zaidi et al (2020) stated that the measurement of band 3 (AE1, SLC4A1, CD233) content of RBCs by eosin-5- maleimide (EMA) staining is swiftly replacing conventional osmotic fragility (OF) test as a tool for laboratory confirmation of HS world-while.  These researchers had systematically evaluated the EMA test as a method to screen for a variety of anemias in the past decade; and compared these findings to those obtained with the OGE (osmoscan), which they had used in the last 30 years.  Their overall experience allowed them to characterize the distinctive patterns with the 2 tests in several congenital erythrocyte membrane disorders, such as HS, HE, Southeast Asian ovalocytosis (SAO), hereditary pyro-poikilocytosis (HPP) variants, erythrocyte volume disorders, various red cell enzymopathies, and hemoglobinopathies.  A crucial difference between the 2 methodologies is that OGE measures RBC deformability of the entire sample of RBCs, while the EMA test examines the band 3 content of individual RBCs.  EMA content is influenced by cell size as smaller RBCs have lower amount of total membrane than larger cells.  The SAO mutation alters the EMA binding site resulting in a lower EMA MCF even as the band 3 content itself is unchanged.  The authors conclude that EMA scan results should be interpreted with caution and both the histograms and dot plots should be analyzed in the context of the clinical picture and morphology.

Berrevoets et al (2021) stated that HS is the most common form of hereditary chronic hemolytic anemia.  It is caused by mutations in RBC membrane and cytoskeletal proteins, which compromise membrane integrity, leading to vesiculation.  This would result in entrapment of poorly deformable spherocytes in the spleen.  Splenectomy is a procedure often carried out in patients with HS. The clinical benefit results from removing the primary site of destruction; thus, improving RBC survival.  However, whether changes in RBC properties contribute to the clinical benefit of splenectomy is unknown.  These researchers employed ektacytometry to examine the longitudinal effects of splenectomy on RBC properties in 5 well-characterized HS patients at 4 different time-points and in a case-control cohort of 26 HS patients; OGE showed that splenectomy resulted in improved intra-cellular viscosity (hydration state) whereas total surface area (TSA) and surface-to-volume ratio remained essentially unchanged.  The cell membrane stability test (CMST), which evaluates the in-vitro response to shear stress, showed that after splenectomy, HS RBCs had partly regained the ability to shed membrane, a property of healthy RBCs, which was confirmed in the case-control cohort.  In particular the CMST holds promise as a novel biomarker in HS that reflects RBC membrane health and may be used to examine treatment response in HS.

Furthermore, an UpToDate review on “Hereditary elliptocytosis and related disorders” (Mentzer, 2021) states that “Osmotic gradient ektacytometry (OGE) measures the deformability and hydration of an RBC population.  OGE is effective in confirming the diagnosis when RBC morphology does not provide a clear diagnosis.  OGE is also helpful in distinguishing HE from hereditary spherocytosis (HS) and hereditary xerocytosis.  OGE can also measure the mechanical stability of RBC ghosts, which has been shown to be abnormal in individuals with HPP”.

RBC Adhesion Index

Dupuy and associates (2019) stated that micro-fluidic devices have become an integral method of cardiovascular research as they enable the study of shear force in biological processes, such as platelet function and thrombus formation.  In addition, micro-fluidic chips offer the benefits of ex-vivo testing of platelet adhesion using small amounts of blood or purified platelets.  Micro-fluidic chips comprise flow channels of varying dimensions and geometries that are connected to a syringe pump.  The pump draws blood or platelet suspensions through the channel(s) allowing for imaging of platelet adhesion and thrombus formation by fluorescence microscopy.  The chips can be fabricated from various blood-compatible materials.  The current protocol uses commercial plastic or in-house polydimethylsiloxane (PDMS) chips.  Commercial biochips offer the advantage of standardization whereas in-house chips offer the advantage of decreased cost and flexibility in design.  The authors concluded that micro-fluidic devices are a powerful tool to study the biorheology of platelets and other cell types with the potential of a diagnostic and monitoring tool for cardiovascular diseases.

Man and colleagues (2020) noted that up-regulated expression of P-selectin on activated endothelium and platelets significantly contributes to the initiation and progression of vaso-occlusive crises (VOC), a major cause of morbidity in sickle cell disease (SCD).  Crizanlizumab (Adakveo), a humanized monoclonal antibody against P-selectin, primarily inhibits the interaction between leukocytes and P-selectin; and has been shown to decrease the frequency of VOCs in clinical trials.  However, the lack of reliable in-vitro assays that objectively measure leukocyte adhesion to P-selectin remains a critical barrier to assessing and improving the therapeutic treatment in SCD.  These researchers presented a standardized micro-fluidic BioChip whole blood adhesion assay to evaluate leukocyte adhesion to P-selectin under physiologic flow conditions.  The findings demonstrated heterogeneous adhesion by leukocytes to immobilized P-selectin, and dose-dependent inhibition of this adhesion following pre-exposure to crizanlizumab.  More importantly, treatment with crizanlizumab following adhesion to P-selectin promoted detachment of rolling, but not of firmly adherent leukocytes.  The authors concluded that these promising findings suggested that the in-vitro microfluidic whole blood adhesion assay, the SCD Biochip, has the potential to enhance the understanding of the underlying mechanisms needed to develop targeted therapeutic strategies during drug development and in clinical trials.  These researchers are currently examining if these assays can predict individual patient response to targeted therapies.

Xuan et al (2018) noted that the immunoassays that are available for the serological diagnosis of the more common subtypes of auto-immune blistering diseases (AIBD) such as pemphigus vulgaris (PV) and pemphigus foliaceus (PF) include enzyme-linked immunosorbent assay (ELISA) testing to specific antigens desmoglein (Dsg)1 and Dsg3, direct immunofluorescence (DIF), indirect immunofluorescence (IIF), and immune-blotting.  These investigators carried out a review of the literature on the BIOCHIP assay.  A total of 6 studies examined the validity of a new BIOCHIP, mosaic-based, IIF test in patients with pemphigus and demonstrated its relatively high sensitivity and specificity (Dsg3: 97.62 to 100 %, 99.6 to 100 %; Dsg1: 90 %, 100 %) in comparison with ELISA (Dsg3: 81 to 100 %, 94 to 100 %; Dsg1: 69 to 100 %, 61.1 to 100 %), and/or IIF (PV: 75 to 100 %, 91.8 to 100 %; PF: 67 to 100 %) using suitable substrates.  So far, validation studies of the BIOCHIP have been carried out in 4 countries (Germany, Italy, Poland, and Turkey) but none in the southern hemisphere.  Caucasian patients were recruited as normal controls for these studies; therefore, the diagnostic value of the BIOCHIP remains uncertain in population groups of other ethnicities.  A range of disease control patients were recruited including patients with linear immunoglobulin A dermatosis, psoriasis, discoid lupus erythematosus, lichen planus, and non-inflammatory skin diseases (e.g., squamous cell carcinoma, basal cell carcinoma, and vascular leg ulcers).  The authors concluded that prospective studies with control patients from a diverse range of ethnicities are needed to better validate the BIOCHIP.  Moreover, these researchers noted that drawbacks of the studies identified in this review included small sample sizes, low case-to-control ratios, and selection bias; they stated that further investigations are needed to address these issues and to validate the use of the BIOCHIP in the diagnosis of pemphigus.

Arunprasath and co-workers (2020) stated that AIBD are a heterogeneous group of diseases characterized by auto-antibodies against desmosomal proteins in the pemphigus group of disorders and adhesion molecules of the dermal-epidermal junction in pemphigoid group of diseases.  DIF establishes the diagnosis of AIBD by demonstrating intercellular deposits of IgG and C3 in case of pemphigus and linear deposits of IgG and C3 along the basement membrane zone (BMZ) in bullous pemphigoid (BP).  BIOCHIP mosaic-based IIF, a novel diagnostic approach employs detection of characteristic staining pattern and target antigens in a single miniature incubation field.  In a cross-sectional study, these researchers compared the BIOCHIP mosaic-based IIF with DIF in the diagnosis of AIBD.  A total of 40 patients of AIBD in the active phase of the disease were included in the study.  Skin biopsy was carried out in these patients for DIF study and serum was subjected to BIOCHIP mosaic-based IIF assay.  The results were then compared.  DIF revealed a diagnosis of pemphigus in 18 patients and BP in 22 patients.  BIOCHIP showed a diagnosis of pemphigus in 18 patients, BP in 18 patients and floor pattern staining in 4 patients, which could be attributed to any of the floor pattern staining subepidermal blistering disease.  The authors concluded that the findings of this study showed that the diagnosis of AIBD by BIOCHIP is showing a statistically significant correlation with that of DIF.  Moreover, these researchers stated that drawbacks of this study included small sample size (n = 40), the lack of a control group and no comparison made with ELISA.

Nili et al (2021) noted that PV is an intra-epidermal AIBD characterized by auto-antibodies against desmosomal adhesion proteins, most commonly Dsg3, leading to the supra-basal cleft formation and acantholysis.  DIF and IIF studies display the intercellular deposition of IgG/C3 throughout the epidermis and presence of circulating auto-antibodies, respectively, as a net-like pattern.  However, the target antigen remains unknown using immune-fluorescence techniques.  Thanks to the development of Dsg ELISA, using recombinant technology, circulating antibodies against Dsg1 and 3 could be detected sensitively.  It is possible to differentiate PV from PF by means of this assay.  BIOCHIP mosaic and multi-variant ELISA are 2 novel serologic methods with the added value of the ability to screen several AIBDs simultaneously.  Non-Dsg1/3 antigens are also involved in the pathogenesis of PV and investigated more deeply thanks to the protein microarrays technique.  furthermore, patients with high values of anti-Dsg1/3 may be lesion-free, suggesting the presence of non-pathogenic auto-antibodies.  The authors concluded that newer diagnostic methods to replace traditional techniques should possess high sensitivity and specificity and be widely available, non-invasive, and relatively cheap.  Moreover, these researchers stated that these newly developed methods need to be further investigated before being recommended for routine use.


References

The above policy is based on the following references:

  1. Albe X, Vassilakos P, Helfer-Guarnori K, et al. Independent prognostic value of ploidy in colorectal cancer: A prospective study using image cytometry. Cancer. 1990;66:1168-1175.  
  2. Alberta Provincial Hematology Tumour Team. Lymphoma. Clinical Practice Guideline No. LYHE-002. Edmonton, AB: Alberta Health Services, Cancer Care; September 2012.
  3. Araujo SE, Bernardo WM, Habr-Gama A, et al. DNA ploidy status and prognosis in colorectal cancer: A meta-analysis of published data. Dis Colon Rectum. 2007;50(11):1800-1810.
  4. Bast RC Jr, Ravdin P, Hayes DF, et al. 2000 update of recommendations for the use of tumor markers in breast and colorectal cancer: Clinical practice guidelines of the American Society of Clinical Oncology. J Clin Oncol. 2001;19(6):1865-1878.
  5. Bauer KD, Bagwell CB, Giaretti W, et al. Consensus review of the clinical utility of DNA flow cytometry in colorectal cancer. Cytometry. 1993;14:486-491.  
  6. Byers CD. Clinical applications of flow cytometry. Clin Lab Sci. 1993;6(3):174-176. 
  7. Camplejohn RS. Flow cytometry. J Pathol. 1992;166(3):323-326.  
  8. Currin SM, Lee SE, Walther PJ. Flow cytometric assessment of deoxyribonucleic acid content in renal adenocarcinoma: Does ploidy status enhance prognostic stratification over stage alone? J Urol. 1990;143:458-463.  
  9. Dabic MM, Nola M, Tomicic I, et al. Adenocarcinoma of the uterine cervix: Prognostic significance of clinicopathologic parameters, flow cytometry analysis and HPV infection. Acta Obstet Gynecol Scand. 2008;87(3):366-372.
  10. Eissa S, Kassim S, El-Ahmady O. Detection of bladder tumours: Role of cytology, morphology-based assays, biochemical and molecular markers. Curr Opin Obstet Gynecol. 2003;15(5):395-403.
  11. Elit L, Fyles A, Chambers A, et al.; Gynecology Cancer Disease Site Group. Adjuvant care for stage I ovarian cancer. Practice Guideline Report #4-13. Cancer Care Ontario Program in Evidence-Based Care. Toronto, ON: Cancer Care Ontario; May 3, 2004.
  12. Gilman-Sachs A. Flow cytometry. Anal Chem. 1994;66(13):700A-707A.
  13. Goetzman EA. Flow cytometry: Basic concepts and clinical applications in immunodiagnostics. Clin Lab Sci. 1993;6(3):177-182.  
  14. Habermehl P, Knuf M, Schwarz M, et al. Flow-cytometric DNA analysis of intracranial tumors in children. Pathol Res Pract. 2004;200(3):197-202.
  15. Harris L, Fritsche H, Mennel R, et al; American Society of Clinical Oncology. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol. 2007;25(33):5287-5312. 
  16. Hedley D. DNA flow cytometry and breast cancer. Breast Cancer Res Treat. 1993;28:51-53.  
  17. Huang HY, Sung MT, Eng HL, et al. Solitary fibrous tumor of the abdominal wall: A report of two cases immunohistochemical, flow cytometric, and ultrastructural studies and literature review. APMIS. 2002;110(3):253-262.
  18. Jackson JF, Boyett J, Pullen J, et al. Favorable prognosis associated with hyperdiploidy in children with acute lymphocytic leukemia correlates with extra chromosome 6: A Pediatric Oncology Group study. Cancer. 1990;66(6):1183-1189.  
  19. Jennings CD, Foon KA. Recent advances in flow cytometry: Application to the diagnosis of hematologic malignancy. Blood. 1997;90(8):2863-2892.  
  20. Jiao YF, Sugai T, Suzuki M, et al. Application of the crypt isolation technique to the flow cytometric analysis of DNA content in gastric carcinoma. Hum Pathol. 2004;35(5):587-593.
  21. Johnson KL. Basics of flow cytometry. Clin Lab Sci. 1992;5(1):22-24.  
  22. Kang YJ, Serhrouchni S, Makhro A, et al. Simple assessment of red blood cell deformability using blood pressure in capillary channels for effective detection of subpopulations in red blood cells. ACS Omega. 2022;7(43):38576-38588.
  23. Kase S, Ishijima K, Uraki T, et al. Usefulness of flow cytometry in diagnosis of IgG4-related ophthalmic disease and extranodal marginal zone B-cell lymphoma of the ocular adnexa. Anticancer Res. 2017;37(9):5001-5004
  24. King TE Jr. Clinical manifestations and diagnosis of pulmonary sarcoidosis. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed January 2015.
  25. Lage JM, Weinberg DS, Huettner PC, et al. Flow cytometric analysis of nuclear DNA content in ovarian tumors: Association of ploidy with tumor type, histologic grade, and clinical stage. Cancer. 1992;69(11)2668-2675.  
  26. Lee SE, Currin SM, Paulson DF, et al. Flow cytometric determination of ploidy in prostatic adenocarcinoma: A comparison with seminal vesicle involvement and histopathological grading as a predictor of clinical recurrence. J Urol. 1988;140(4):769-774.  
  27. Locker GY, Hamilton S, Harris J, et al. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol. 2006;24(33):5313-5327.
  28. Lu Y, Liang H, Yu T, et al. Isolation and characterization of living circulating tumor cells in patients by immunomagnetic negative enrichment coupled with flow cytometry. Cancer. 2015;121(17):3036-3045.
  29. Ludovini V, Pistola L, Gregorc V, et al. Biological markers and DNA flow cytometric analysis in radically resected patients with non-small cell lung cancer. A study of the Perugia Multidisciplinary Team for Thoracic Tumors. Tumori. 2008;94(3):398-405.
  30. Martinez JE, Beck JR, Allsbrook WC Jr., et al. Flow cytometric DNA analysis. Clin Lab Sci. 1990;3(3):180-183.
  31. Mesiwala AH, Scampavia LD, Rabinovitch PS, On-line flow cytometry for real-time surgical guidance. Neurosurgery. 2004;55(3):551-560; discussion 560-561.
  32. Midthun DE. Overview of the initial evaluation, treatment and prognosis of lung cancer. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed January 2016.
  33. Milligan DW, Grimwade D, Cullis JO, et al. Guidelines on the management of acute myeloid leukaemia in adults. London, UK: British Society of Haematology (BSH); May 23, 2005.
  34. Minnema MC, Kimby E, D'Sa S, et al. Guideline for the diagnosis, treatment and response criteria for Bing-Neel syndrome. Haematologica. 2017;102(1):43-51.
  35. Mirza AN, Mirza NQ, Vlastos G, Singletary SE. Prognostic factors in node-negative breast cancer: A review of studies with sample size more than 200 and follow-up more than 5 years. Ann Surg. 2002;235(1):10-26.
  36. Nardo-Marino A, Glenthøj A, Brewin JN, et al. The significance of spleen size in children with sickle cell anemia. Am J Hematol. 2022;97(12):1520-1528.
  37. National Comprehensive Cancer Network (NCCN). Myeloproliferative neoplasms. NCCN Clinical Practice Guidelines in Oncology, Version 2.2017. Fort Washington, PA: NCCN; 2017.
  38. National Comprehensive Cancer Network (NCCN). NCCN Biomarkers Compendium. Fort Washington, PA: NCCN; 2017. 
  39. National Comprehensive Cancer Network (NCCN). Small cell lung cancer. NCCN Clinical Practice Guidelines in Oncology, Version 1.2016. Fort Washington, PA: NCCN; 2016.
  40. National Health and Medical Research Council (NHMRC). Clinical Practice Guidelines: The management of cutaneous melanoma. A report of the Australian Cancer Network Working Party on Cutaneous Melanoma. Canberra, ACT: AusInfo Government Info; December 1999.
  41. National Health and Medical Research Council (NHMRC). Clinical Practice Guidelines: Evidence-based information and recommendations for the management of localised prostate cancer. A report of the Australian Cancer Network Working Party on Management of Localised Prostate Cancer. Canberra, ACT: AusInfo Government Info; 2003.
  42. Nativ O, Winkler HZ, Raz Y, et al. Stage C prostatic adenocarcinoma: Flow cytometric nuclear DNA ploidy analysis. Mayo Clinic Proc. 1989;64(8):911-919.  
  43. No authors listed. Statement on sarcoidosis. Joint Statement of the American Thoracic Society (ATS), the European Respiratory Society (ERS) and the World Association of Sarcoidosis and Other Granulomatous Disorders (WASOG) adopted by the ATS Board of Directors and by the ERS Executive Committee, February 1999. Am J Respir Crit Care Med. 1999;160(2):736-755.
  44. Otchy D, Hyman NH, Simmang C, et al. Practice parameters for colon cancer. Dis Colon Rectum. 2004;47(8):1269-1284.
  45. Ottesen GL. Carcinoma in situ of the female breast. A clinico-pathological, immunohistological, and DNA ploidy study. APMIS Suppl. 2003;(108):1-67.
  46. Pisani T, De Iorio P, Cenci M, et al. Evaluation of cellular proliferative activity and DNA ploidy as a prognostic factor in breast cancer. Minerva Ginecol. 1999;51(1-2):1-5.  
  47. Rew DA. Clinical applications of flow cytometry. Br J Hosp Med. 1992;48(3-4):171-175.
  48. Ritchie AW, Dorey F, Layfield LJ, et al. Relationship of DNA content to conventional prognostic factors in clinically localised carcinoma of the prostate. B J Urol. 1988;62(3):254-260.  
  49. Royal College of Obstetricians and Gynaecologists (RCOG). The management of gestational trophoblastic neoplasia. Guideline No. 38. London, UK: RCOG; February 2004.
  50. Russo A, Bazan V, Migliavacca M, et al. DNA aneuploidy and high proliferative activity but not K-ras-2 mutations as independent predictors of clinical outcome in operable gastric carcinoma: Results of a 5-year Gruppo Oncologico dell'Italia Meridonale (GDIM) prospective study. Cancer. 2001;92(2):294-302.
  51. Shearer WT, Buckley RH, Engler RJM, et al. Practice parameters for the diagnosis and management of immunodeficiency. Ann Allergy Asthma Immunol. 1996;76(3):282-294.  
  52. Shelley W, Trudeau M; Breast Cancer Disease Site Group. Adjuvant systemic therapy for node-negative breast cancer. Practice Guideline Report #1-8. Cancer Care Ontario Program in Evidence-Based Care. Toronto, ON: Cancer Care Ontario; February 2002.
  53. Silvestrini R, D'Agnano I, Faranda A, et al. Flow cytometric analysis of ploidy in colorectal cancer: A multicentric experience. Br J Cancer. 1993;67:1042-1046.  
  54. Smith A, Wisloff F, Samson D; UK Myeloma Forum, Nordic Myeloma Study Group, British Committee for Standards in Haematology. Guidelines on the diagnosis and management of multiple myeloma 2005. Br J Haematol. 2006;132(4):410-451.
  55. Suehiro Y, Okada T, Okada T, et al. Aneuploidy predicts outcome in patients with endometrial carcinoma and is related to lack of CDH13 hypermethylation. Clin Cancer Res. 2008;14(11):3354-3361.
  56. Sutcliffe P, Hummel S, Simpson E, et al. Use of classical and novel biomarkers as prognostic risk factors for localised prostate cancer: A systematic review. Health Technol Assess. 2009;13(5):1-242.
  57. Tomoda H, Kakeji Y, Furusawa M. Prognostic significance of flow cytometric analysis of DNA content in colorectal cancer: A prospective study. J Surg Onc. 1993;53:144-148.  
  58. Trope C, Kaern J. DNA ploidy in epithelial ovarian cancer: A new independent prognostic factor? Gynecol Oncol. 1994;53:1-4.
  59. Villas BH. Flow cytometry: An overview. Cell Vis. 1998;5(1):56-61.
  60. Winchester DP. Standards for diagnosis and management of invasive breast carcinoma. American College of Radiology. American College of Surgeons. College of American Pathologists. Society of Surgical Oncology. CA Cancer J Clin. 1998;48(2):83-107.  
  61. Wolfson AH, Winter K, Crook W, et al. Are increased tumor aneuploidy and heightened cell proliferation along with heterogeneity associated with patient outcome for carcinomas of the uterine cervix? A combined analysis of subjects treated in RTOG 9001 and a single-institution trial. Int J Radiat Oncol Biol Phys. 2008;70(1):111-117.
  62. Yasa MH, Bektas A, Yukselen V, et al. DNA analysis and DNA ploidy in gastric cancer and gastric precancerous lesions. Int J Clin Pract. 2005;59(9):1029-1033.
  63. Zhang Z, Weaver DL, Munjal K, Evans MF. Intratumoral DNA content heterogeneity in breast carcinomas demonstrated by core punch tissue sampling and flow cytometry. J Clin Pathol. 2014;67(9):821-824.

Osmotic Gradient Ektacytometry

  1. Berrevoets MC, Bos J, Huisjes R, et al. Ektacytometry analysis of post-splenectomy red blood cell properties identifies cell membrane stability test as a novel biomarker of membrane health in hereditary spherocytosis. Front Physiol. 2021;12:641384.
  2. Huisjes R, Makhro A, Llaudet-Planas E, et al. Density, heterogeneity and deformability of red cells as markers of clinical severity in hereditary spherocytosis. Haematologica. 2020;105(2):338-347.
  3. Llaudet-Planas E, Vives-Corrons JL, Rizzuto V, et al. Osmotic gradient ektacytometry: A valuable screening test for hereditary spherocytosis and other red blood cell membrane disorders. Int J Lab Hematol. 2018;40(1):94-102.
  4. Mentzer WC. Hereditary elliptocytosis and related disorders. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed December 2021.
  5. Parrow NL, Violet P-C, Tu H, et al. Measuring deformability and red cell heterogeneity in blood by ektacytometry. J Vis Exp. 2018;(131):56910.
  6. Zaidi AU, Buck S, Gadgeel M, et al. Clinical diagnosis of red cell membrane disorders: Comparison of osmotic gradient ektacytometry and eosin maleimide (EMA) fluorescence test for red cell band 3 (AE1, SLC4A1) content for clinical diagnosis. Front Physiol. 2020;11:636.

RBC Adhesion Index

  1. Arunprasath P, Rai R, Venkataswamy C, et al. Comparative analysis of BIOCHIP mosaic-based indirect immunofluorescence with direct immunofluorescence in diagnosis of autoimmune bullous diseases: A cross-sectional study. Indian Dermatol Online J. 2020;11(6):915-919.
  2. Dupuy A, Ju LA , Passam FH, et al. Straight channel microfluidic chips for the study of platelet adhesion under flow. Bio Protoc. 2019;9(6):e3195.
  3. Man Y, Goreke U, Kucukal E, et al. Leukocyte adhesion to P-selectin and the inhibitory role of Crizanlizumab in sickle cell disease: A standardized microfluidic assessment. Blood Cells Mol Dis. 2020;83:102424.
  4. Nili A, Farid AS, Asgari M, et al. Current status and prospects for the diagnosis of pemphigus vulgaris. Expert Rev Clin Immunol. 2021;17(8):819-834.
  5. Xuan RR, Yang A, Murrell DF, et al. New biochip immunofluorescence test for the serological diagnosis of pemphigus vulgaris and foliaceus: A review of the literature. Int J Womens Dermatol. 2018;4(2):102-108.